Feeds:
Posts
Comments

Posts Tagged ‘retinoids’

Vitamin D debates

Larry H. Bernstein, MD, FCAP, Curator

LPBI

 

Vitamin D: Time for Rational Decision-Making

JoAnn E. Manson, MD, DrPH

Hello. This is Dr JoAnn Manson, professor of medicine at Harvard Medical School and Brigham and Women’s Hospital. I would like to talk with you about the vitamin D dilemma. The question of whether to screen routinely for vitamin D deficiency or to recommend high-dose vitamin D supplementation for our patients continues to be one of the most perplexing and vexing issues in clinical practice, and many clinicians are seeking guidance on these issues.

There appears to be a growing disconnect between the observational studies and the randomized clinical trials of vitamin D. For example, the observational studies are showing a fairly consistent relationship between low blood levels of vitamin D and an increased risk for heart disease, cancer, diabetes, and many other chronic diseases. Yet, the randomized clinical trials of vitamin D supplementation to date have been generally disappointing. This includes several randomized trials published over the past few months, including a meta-analysis[1] of randomized trials of vitamin D supplementation showing minimal, if any, benefit in terms of lowering blood pressure; a trial[2] of high-dose vitamin D supplementation showing no clear benefit for muscle strength, bone mineral density, or even the risk for falls; and, most recently, a randomized trial[3] of vitamin D supplementation with and without calcium showing no clear benefit in reducing the risk for colorectal adenomas. The latter trial was very recently published in the New England Journal of Medicine.

The Institute of Medicine (IOM)[4] and the US Preventive Services Task Force[5] do not endorse routine universal screening for vitamin D deficiency. They also recommend more moderate intakes [of vitamin D]. For example, the IOM recommends 600-800 IU a day for adults and also recommends avoiding daily intakes above 4000 IU, which has been set as the tolerable upper intake level.

However, it is important to keep in mind that these are public health population guidelines for a generally healthy population, and they by no means preclude individual decision-making by the clinician in the context of a patient who may have health conditions or risk factors that would indicate a benefit from targeted screening for vitamin D deficiency or higher-dose supplementation. For example, some patients may have higher vitamin D requirements. This may include patients with bone health problems (osteoporosis, osteomalacia) or poor diets, those who spend minimal time outdoors, those with malabsorption syndromes, or those who take medications that may interfere with vitamin D metabolism (glucocorticoids, anticonvulsant medications, and antituberculosis drugs). Therefore, overall, there is a role for individualized decision-making, in terms of screening for vitamin D deficiency in patients who have bone health problems or special risk factors, and even treating with higher doses of vitamin D, which may go above 4000 IU a day in patients who have higher requirements.

In the next several years, large-scale, randomized trials of vitamin D supplementation, including high-dose vitamin D supplementation, will be completed—and these results will be published. They will help to inform clinical decision-making, so stay tuned for those results.

Thank you so much for your attention. This is JoAnn Manson.

References

  1. Beveridge LA, Struthers AD, Khan F, et al. D-PRESSURE Collaboration. Effect of vitamin D supplementation on blood pressure: a systematic review and meta-analysis incorporating individual patient data. JAMA Intern Med. 2015;175:745-754. Abstract
  2. Hansen KE, Johnson RE, Chambers KR, et al. Treatment of vitamin D insufficiency in postmenopausal women: a randomized clinical trial. JAMA Intern Med. 2015;175:1612-1621. Abstract
  3. Baron JA, Barry EL, Mott LA, et al. A trial of calcium and vitamin D for the prevention of colorectal adenomas. N Engl J Med. 2015;373:1519-1530. Abstract
  4. Institute of Medicine. Dietary Reference Intakes for Calcium and Vitamin D. Washington, DC: National Academies Press; 2011. http://iom.nationalacademies.org/Reports/2010/Dietary-Reference-Intakes-for-Calcium-and-Vitamin-D.aspx Accessed October 28, 2015.
  5. US Preventive Services Task Force. Final Recommendation Statement: Vitamin D Deficiency: Screening, 2014.http://www.uspreventiveservicestaskforce.org/Page/Document/RecommendationStatementFinal/vitamin-d-deficiency-screening Accessed October 28, 2015.

 

Isn’t there much more to this than the debates entail?

The vitamin D hormone and its nuclear receptor: molecular actions and disease states.
 J Endocrinol. 1997 Sep;154 Suppl:S57-73.      http://dx.doi.org:/10.1677/joe.0.154S057

Vitamin D plays a major role in bone mineral homeostasis by promoting the transport of calcium and phosphate to ensure that the blood levels of these ions are sufficient for the normal mineralization of type I collagen matrix in the skeleton. In contrast to classic vitamin D-deficiency rickets, a number of vitamin D-resistant rachitic syndromes are caused by acquired and hereditary defects in the metabolic activation of the vitamin to its hormonal form, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), or in the subsequent functions of the hormone in target cells. The actions of 1,25(OH)2D3 are mediated by the nuclear vitamin D receptor (VDR), a phosphoprotein which binds the hormone with-high affinity and regulates the expression of genes via zinc finger-mediated DNA binding and protein-protein interactions. In hereditary hypocalcemic vitamin D-resistant rickets (HVDRR), natural mutations in human VDR that confer patients with tissue insensitivity to 1,25(OH)2D3 are particularly instructive in revealing VDR structure function relationships. These mutations fall into three categories: (i) DNA binding/nuclear localization, (ii) hormone binding and (iii) heterodimerization with retinoid X receptors (RXRs). That all three classes of VDR mutations generate the HVDRR phenotype is consistent with a basic model of the active receptor as a DNA-bound, 1,25(OH)2D3-liganded heterodimer of VDR and RXR. Vitamin D responsive elements (VDREs) consisting of direct hexanucleotide repeats with a spacer of three nucleotides have been identified in the promoter regions of positively controlled genes expressed in bone, such as osteocalcin, osteopontin, beta 3-integrin and vitamin D 24-OHase. The 1,25(OH)2D3 ligand promotes VDR-RXR heterodimerization and specific, high affinity VDRE binding, whereas the ligand for RXR, 9-cis retinoic acid (9-cis RA), is capable of suppressing 1,25(OH)2D3-stimulated transcription by diverting RXR to form homodimers. However, initial 1,25(OH)2D3 liganding of a VDR monomer renders it competent not only to recruit RXR into a heterodimer but also to conformationally silence the ability of its RXR partner to bind 9-cis RA and dissociate the heterodimer. Additional probing of protein-protein interactions has revealed that VDR also binds to basal transcription factor IIB (TFIIB) and, in the presence of 1,25(OH)2D3, an RXR-VDR-TFIIB ternary complex can be created in solution. Moreover, for transcriptional activation by 1,25(OH)2D3, both VDR and RXR require an intact short amphipathic alpha-helix, known as AF-2, positioned at their extreme C-termini. Because the AF-2 domains participate neither in VDR-RXR heterodimerization nor in TFIIB association, it is hypothesized that they contact, in a ligand-dependent fashion, transcriptional coactivators such as those of the steroid receptor coactivator family, constituting yet a third protein-protein interaction for VDR. Therefore, in VDR-mediated transcriptional activation, 1,25(OH)2D3 binding to VDR alters the conformation of the ligand binding domain such that it: (i) engages in strong heterodimerization with RXR to facilitate VDRE binding, (ii) influences the RXR ligand binding domain such that it is resistant to the binding of 9-cis RA but active in recruiting coactivator to its AF-2 and (iii) presents the AF-2 region in VDR for coactivator association. The above events, including bridging by coactivators to the TATA binding protein and associated factors, may position VDR such that it is able to attract TFIIB and the balance of the RNA polymerase II transcription machinery, culminating in repeated transcriptional initiation of VDRE-containing, vitamin D target genes. Such a model would explain the action of 1,25(OH)2D3 to elicit bone remodeling by stimulating osteoblast and osteoclast precursor gene expression, while concomitantly triggering the termination of its hormonal signal by inducing the 24-OHase catabolizing enzyme.

 

Classic nutritional rickets is caused by the simultaneous deprivation of sunlight exposure and dietary vitamin D. As depicted in Fig. 1, the pathways comprising the metabolic activation of the vitamin to its hormonal form and subsequent functions in target tissues present a number of additional steps where defects elicit vitamin D-resistant rachitic syndromes. Two of these disorders involve the inadequate bioactivation of 25-hydroxy¬ vitamin D3 (25(OH)D3) to 1,25-dihydroxyvitamin D3 (l,25(OH)2D3) by the kidney as catalyzed by the 1-OHase enzyme (Fig. 1).

Figure 1 Bioactivation of vitamin D3 and actions of the 1,25(OH)2D3 hormonal metabolite on intestine, bone and kidney, along with related rachitic syndromes. The production of 1,25(OH)2D3 is depicted in the lowet portion and its functions on mineral ttansport in target cells ate pictured in the upper portion. Defects eliciting rachitic syndromes ate boxed, with the televant mutated gene and chromosomal location denoted where appropriate

Acquired chronic renal failure results in renal rickets and secondary hyperparathyroidism (renal osteodystrophy) when the compromising of renal mass reduces 1-OHase activity (Haussier oc McCain 1977). The etiology of pseudo-vitamin D-deficiency rickets (PDDR) apparently involves a hereditary defect in the gene coding for the 1-OHase enzyme (Labuda et al. 1992). Interestingly, the PDDR locus is resolvable from that of the vitamin D receptor (VDR) but maps very close to it on chromosome 12 in the 12ql3—14 region (Labuda et al. 1992). Recently, a cDNA was cloned for the rat 1-OHase (St-Arnaud et al. 1996) and it is expected that the human renal 1-OHase gene will soon be cloned and its chromosomal location determined. The likelihood that both the gene encoding the enzyme that generates the l,25(OH)2D3 hormone and the cognate hormone receptor gene lie in close proximity on chromosome 12 invites speculation about the evolution of the vitamin D ligand receptor system. The traditional actions of vitamin D, via its l,25(OH)2D3 hormonal metabolite, are to effect calcium and phosphate homeostasis to ensure the deposition of bone mineral on type I collagen matrix (summarized in Fig. 1).

Figure 1 Bioactivation of vitamin D3 and actions of the 1,25(OH)2D3 hormonal metabolite on intestine, bone and kidney, along with related rachitic syndromes. The production of 1,25(OH)2D3 is depicted in the lowet portion and its functions on mineral ttansport in target cells ate pictured in the upper portion. Defects eliciting rachitic syndromes ate boxed, with the televant mutated gene and chromosomal location denoted where appropriate

 

l,25(OH)2D3 stimulates intestinal calcium and phosphate absorption, bone calcium and phosphate résorption, and renal calcium and phosphate reabsorption, all resulting in a sufficient CaP04 ion product to precipitate hydroxyapatite. Failure to achieve normal bone mineral accretion by these mechanisms leads to rachitic syndromes. Recently, a breakthrough has occurred in our understand¬ ing of what was originally known as hypophosphatemic vitamin D-resistant rickets, a familial disorder of renal phosphate wasting more appropriately referred to as dominant X-linked hypophosphatemic (HYP) rickets (Fig. 1). The gene defect responsible for HYP rickets has been fine mapped in the Xp22T region, harboring a gene identified as PEX, or phosphate regulating gene with homologies to endopeptidases located on the X-chromosome (Francis et al. 1995). One hypothesis is that PEX codes for an endopeptidase that apparently correctly processes a peptide precursor to yield a novel, as yet unidentified, phosphate retaining hormone. The normal function of this hormone may be to oppose the action of parathyroid hormone (PTH) and stimulate phosphate reabsorption by the renal tubule by inducing the Na -phosphate cotransporter. However, the existence of tumor-induced osteomalacia, an acquired disorder that closely resembles the phosphate wasting of HYP rickets and is characterized by low circulating l,25(OH)2D3 (Parker et al. 1981), combined with renal cross-transplantation (Nesbitt et al. 1992) and parabiosis (Meyer et al. 1989) studies in normal and hyp mice, indicates strongly that the HYP phenotype is caused by excessive amounts of a phosphaturic hormone in the circulation. This humoral peptide is distinct from PTH and has been named phosphatonin (Cai et al. 199A, Econs & Drezner 1994). Thus, instead of PEX mutations result¬ ing in insufficient generation of a novel phosphate retaining peptide, they may instead elicit the appearance of abnormally high circulating levels of phosphatonin, with the normal role of the PEX gene product postulated to be the proteolytic inactivation of this phosphaturic principle. Most germane to the vitamin D endocrine system is the fact that serum l,25(OH)2D3 levels are inappropriately low for the prevailing phosphate concentrations in HYP rickets and patients can be cured with a therapeutic combination of phosphate and l,25(OH)2D3 (Harrel et al. 1985). Because it is well known that hypophosphatemia stimulates l,25(OH)2D3 production (Hughes et al. 1975), the PEX/phosphatonin system might constitute yet another regulatory loop in maintaining normal phosphate homeostasis. One could hypothesize that under hypo- phosphatemic conditions, when l,25(OH)2D3 levels are elevated, the sterol hormone not only increases intestinal phosphate absorption (Fig. 1) and suppresses PTH synthesis (DeMay et al. 1992) to conserve phosphate, but also induces the PEX gene product (Rowe et al. 1996) to cleave phosphatonin and further promote renal phosphate reclamation. l,25(OH)2D3 is primarily recognized as a calcémic hormone, perhaps due to the abundance of dietary phosphate, or because calcium homeostasis is more vitamin D-dependent than the regulation of extracellular phos¬ phate. Regardless of the mechanism, traditional vitamin D-deficiency and clinically significant defects in the vitamin D receptor lead invariably to hypocalcemia and secondary hyperparathyroidism, with phosphate being somewhat less affected. As illustrated in Fig. 1, target tissue insensitivity to l,25(OH)2D3 is known as hereditary hypocalcémie vitamin D-resistant rickets (HVDRR) and is caused by defects in the gene on chromosome 12 coding for the VDR. A review of the etiology of HVDRR and the natural mutations in the VDR that confer tissue insensitivity and clinical resistance to l,25(OH)2D3 is particularly instructive in illuminating the physiologic relevance of the l,25(OH)-,D3-VDR hormone-receptor complex as well as structure/function relationships in the receptor itself.

Natural mutations in the nuclear vitamin D receptor Clinically significant hereditary hypocalcémie vitamin D-resistant rickets is an autosomal recessive disorder resulting in a phenotype characterized by severe bowing of the lower extremities, short stature and, often, alopecia (Rut et al. 199A). The serum chemistry in HVDRR includes frank hypocalcemia, secondary hyperpara¬ thyroidism, elevated alkaline phosphatase, variable hypophosphatemia and markedly increased l,25(OH)2D3. The symptoms of HVDRR, with the exception of alopecia, mimic classic vitamin D-deficiency rickets, suggesting that VDR not only mediates the bone mineral homeostatic actions of vitamin D but may also participate in the differentiation of hair follicles in utero. Recently, VDR knockout mice have been created (Yoshizawa et al. 1996), revealing apparently normal hétérozygotes but severely affected homozygotes (VDR-/-), 90% ofwhich die within 8—10 weeks. Surviving mice lose their hair and possess low bone mass, hypocalcemia, hypophosphatemia and 10-fold elevated l,25(OH)2D3 coincident with extremely low 24,25(OH)2D3. All of these parameters in the VDR knockout mouse mimic the phenotype of patients with HVDRR, confirming that VDR normally mediates all of the bone mineral regulating functions of vitamin D. Interestingly, although natural point mutations in other receptors related to VDR, such as thyroid hormone receptor ß (TRß) (Collingwood et al. 1994), are charac¬ terized by dominant negative receptors that generate the thyroid hormone resistant phenotype in the heterozygotic context, no natural, dominant negative mutations have yet been identified in HVDRR patients (Whitfield et al. 1996). Thus, all HVDRR cases studied to date are homozygous for the particular VDR mutation.

Figure 2 Natural mutations in the human vitamin D receptor leading to 1,25(OH)2D¡ hormone resistance. See text for details and citations. N37, K91 and E92 are not sites of VDR natural mutations, but are so designated because they ate heterodimerization contacts that lie within the DNA binding domain (Hsieh et al. 1995, Rastinejad et al. 1995). The eight cysteine residues (C) that tetrahedrally coordinate two zinc atoms in the finger sttucture are also denoted.

Figure 2 illustrates a number of point mutations in VDR that have been detected in HVDRR patients (reviewed in Rut et al. 199A, Haussler et al. 1995). Three of these genetic alterations result in nonsense mutations that introduce stop codons in VDR (K73stop, Q152sfo|> and Y295stop), creating truncated VDRs that lack both hormone- and DNA-binding (heterodimerization) capacities and are associated with unstable mRNAs. More revealing are the series of missense mutations (Fig. 2) that can be classified according to three of the basic molecular functions of VDR: (i) DNA binding/nuclear localization by the N-terminal zinc finger region, (ii) l,25(OH)2D3 hormone binding by the C-terminal domain and (iii) heterodimerization with retinoid X receptors (RXRs) through subregions of the C-terminal domain. As depicted schematically in Fig. 2 and discussed in detail later, VDR is a ligand-dependent transcription factor that controls gene expression by heterodimerizing with RXR and associating specifically with vitamin D responsive elements (VDREs) in target genes. Since VDR is a member of the steroid, retinoid, thyroid hormone receptor superfamily, and belongs to the VDR/retinoic acid receptor (RAR)/TR subfamily of RXR heterodimerizing species (Haussler et al. 1991), it is reasonable to draw from data on RAR and TR for comparison with VDR.

The greatest number of VDR natural mutations char¬ acterized to date are localized to the DNA binding, zinc finger region (Fig. 2). The first two discovered, G33D and R73Q (Hughes et al. 1988), reside at the ‘tips’ of the fingers and affect charge—charge interactions between VDR and the phosphate backbone of DNA. When viewed in toto, the zinc finger region mutations in HVDRR (Fig. 2) have the following two general prop¬ erties: (i) they occur in residues conserved across the entire nuclear receptor superfamily and (ii) most lie within -helices on the C-terminal side of the first and second fingers which are intimately involved in DNA base recognition and phosphate backbone contacts respectively (Rastinejad et al. 1995). These observations suggest that many of the clinically significant mutations in VDR which are still compatible with life may not greatly perturb the fundamental structure of the DNA binding domain of the receptor, but instead compromise its ability to recog¬ nize DNA with specificity and high affinity. Whether HVDRR cases with mutations in zinc finger region residues unique to VDR will be uncovered depends upon the properties of such alterations, which could range from innocuous to lethal.

Mutations located within the hormone binding domain of VDR also elicit the HVDRR phenotype (Fig. 2), including R274L (Kristjansson et al. 1993) and H305Q (Malloy et al 1995). Transcriptional activation by R274L and H305G VDR is attenuated as a result of inefficient l,25(OH)2D3 binding, ranging from severe in the case of R274L to a modest increase in Kd for H305Q. In both instances, transcriptional activation is restored when the dose of l,25(OH)2D3 is raised to pharmacologie levels (10 m) in transfection experiments (Kristjansson et al. 1993, Malloy et al. 1995). Our laboratory has recently characterized two novel VDR hormone binding domain mutations in HVDRR patients, I314S and R391C, that significantly affect the heterodimerization of VDR with RXR (Whitfield et al. 1996). Both of these C-terminal replacements (Fig. 2), however, do display some degree of what may be a hormone binding deficit, a phenomenon not observable in typical in vitro ligand binding kinetic assays at 4 °C. Thus, only at 37 °C in intact cells do R391C and I314S exhibit apparent slight and significant impairment of l,25(OH)2D3 high affinity retention respectively (Whitfield et al. 1996). Further, the two mutations in question are situated in or adjacent to heptad repeats (Fig. 2), hypothetical coiled-coil-like structures that were originally proposed to participate in the heterodimerization of VDR, RAR, and TR with RXR (Forman & Samuels 1990, Nakajima et al. 1994). Consist¬ ent with this concept, both R391C and I314S VDRs do not bind RXR with normal affinity when assayed in vitro, with the greatest impairment of heterodimerization occur¬ ring with R391C (affinity reduced by one order of magnitude) (Whitfield et al. 1996). Additional evidence supporting blunted RXR heterodimerization by these two mutant VDRs is provided by transfection experiments in restored to that of normal fibroblasts when fibroblasts from patients harboring either the R391C or the I314S mutation are cotransfected with exogenous RXR. Yet this apparent RXR rescue of the mutated VDRs requires approximately 10-fold elevated l,25(OH)2D3 doses com¬ pared with the response to hormone in normal fibroblasts (Whitfield et al. 1996). This latter observation reveals that the hormone binding and heterodimerization functions of VDR are not entirely separable, an aspect which is also apparent from fundamental biochemical analysis of the hormone dependency of VDR-RXR heterodimer binding to VDREs as discussed in detail below.

Understanding the molecular properties of natural VDR mutations in HVDRR allows us to comprehend why the patients respond differentially to therapy with massive doses of l,25(OH)2D3, or suitable analogs. For example, cases with zinc finger region aberrations are unresponsive to the hormone because DNA binding is precluded by the absence of structural complemen¬ tarity between VDR and the VDRE, regardless of the l,25(OH)2D3 liganding or heterodimerization of the receptor in solution. Conversely, patients harboring mutations in the hormone binding/heterodimerization domain can be responsive to pharmacologie doses of l,25(OH)2D3 or analogs, even though the hormone already is increased in the circulation because of the hypocalcemia caused by tissue insensitivity. For example, patient I314S was essentially cured by excess vitamin D metabolite, indicating that compensating for the hormone binding deficit was able to override the milder heterodimerization defect and allow sufficient VDRE binding by the VDR-RXR heterodimer. Conversely, patient R391C responded only modestly to treatment with excess l,25(OH)2D3 analog, presumably because the fundamental heterodimerization defect could not be overcome and therefore normal VDRE binding could not be achieved (Whitfield et al. 1996).

The final insights gained from the natural VDR mutations summarized in Fig. 2 are structural in nature. We have discussed previously that the zinc finger mutations are confined to absolutely conserved residues. In the crystal structure of the DNA binding domain heterodimers of RXRa and TRß (Rastinejad et al. 1995), the lysine and arginine residues corresponding to K45 and R50 in human VDR (hVDR) make direct base contacts with DNA, while the arginines corresponding to R73 and R80 in hVDR make direct DNA phosphate backbone contacts. That mutations in these four residues are clinically important in the etiology of HVDRR argues for structural congruity between the VDR finger region and that of TR. Rastinejad et al. (1995) have extended this assumption to include a modeling of RXR-TR vs RXRVDR bound to DNA which accommodates the fact that TR binds as a heterodimer to a direct hexanucleotide repeat spaced by four nucleotides (DR+4), while VDR binds as a heterodimer to a similar set of half elements spaced by three nucleotides (DR+3). In addition to verifying the common protein-DNA interfaces, their modeling predicts that hVDR residues N37 in the first finger and K91/E92 C-terminal of the second finger (see Fig. 2) engage in heterodimeric contacts with residues in the second zinc finger ofRXR to form effectively a stable, DNA-supported heterodimer. Indeed, recent site-directed mutational studies (Hsieh et al. 1995) indicate that the alteration ofK91 and E92 in hVDR in fact grossly reduces transactivation while moderately attenuating hetero¬ dimerization and DNA binding, thus confirming the importance of K91 and E92. An additional surprising finding was that the K91/E92 double mutant manifested dominant negative characteristics (Hsieh et al. 1995), distinguishing it from the natural HVDRR replacements discussed above. Apparently, the K91/E92 mutant VDR is able to bind DNA sufficiently through its native zinc finger and strong heterodimerization function in the ligand binding domain such that it can block binding by wild type receptor, but is rendered inactive in stimulating transcription because of a presumed conformational per¬ turbation initiated by unstable or improper alignment of the heterodimer on the VDRE.

Based upon recently reported X-ray crystal structures of the ligand binding domains of ligand-occupied hRARy (Renaud et al. 1995), agonist-occupied rat TRa, (Wagner et al. 1995) and unoccupied, but dimeric hRXRa (Bourguet et al. 1995), it is also possible to incorporate the HVDRR mutations in the hormone binding domain (Fig. 2) into a hypothetical structural context. Figure 3 constitutes a schematic compilation of the existing crystallographic data and compares them with natural and artificially generated mutations in hVDR. At the top of Fig. 3, the residue numbers for VDR in the ligand binding domain appear in relation to the older heptad repeat nomenclature (heptads 1—9, dotted boxes). At least some of these heptads, particularly heptads 4 and 9, are thought to facilitate heterodimerization (Nakajima et al. 1994). The El region is a highly conserved area that supports heterodimerization (Whitfield et al. 1995è). The helices depicted schematically in Fig. 3 (open boxes) are those determined for hRARy; this general pattern of -helices and ß-strands (solid boxes) appears to be well conserved across the TR, RAR and RXR members of the subfamily crystallized thus far (Bourguet et al. 1995, Renaud et al. 1995, Wagner et al. 1995). Although the heterodimerization domains have yet to be elucidated by structural analysis, the homodimerization domain of RXR is comprised of helices 7, 9 and 10 (Fig. 3 and Bourguet et al. 1995). Flanking the dimerization region are clusters of ligand binding contacts, shown for RAR and TR in Fig. 3, which paint a picture of hormone binding involving helices 3, 5, 11 and 12 plus portions of helices 6 and 7 along with their intervening loop, as well as the loop between ß-strands 1 and 2.

Figure 3 Hormone binding (R274L and H305Q) and heterodimerization (I314S and R391C) natural mutations in VDR that confer the HVDRR phenotype are positioned in the context of retinoid and thyroid hormone receptor subfamily ligand binding domain structures. See text for details and citations.

As summarized in Fig. 3 and discussed by Whitfield et al. (1995a, 1996), a number of artificially generated mutants in hVDR support the con¬ cept that the dimerization and honnone binding regions in VDR are well aligned with those in RXR, RAR and TR. Of even greater interest and relevance to the present monograph, the four clinically important hVDR mutants under consideration correspond to pertinent locations in the known structures of the retinoid and thyroid hormone receptor ligand binding domains. We postulate that this general structural organization represents that of the VDR ligand binding domain. As shown in Fig. 3, the pure hormone binding mutant hVDRs, namely R274L and H305Q, are located precisely within ligand clusters in helix 5 and in the loop between helix 6 and 7 respectively. I314S, which endows hVDR with combined defects in hormone retention and heterodimerization, lies within helix 7 at a presumed interface of ligand binding and dimerization activities of the receptor (Fig. 3). Finally, R391C is positioned well within the helix 10 dimerization surface, but not far removed from C-terminal ligand binding contacts that are likely influenced by replacement of this amino acid in hVDR. Thus, at least within the context of the assumed structural organization of VDR derived from that of other subfamily members, the I314S and R391C mutations are situated precisely where they would be predicted to lie, given the biological properties of the mutant receptors and the phenotype of the patients. These results not only have profound implications con¬ cerning the putative structure of VDR in relation to its closest relatives, but prove unequivocally that the calcémic actions of l,25(OH)2D3 are mediated by the vitamin D receptor, existing as a l,25(OH)2D3-liganded heterodimer with RXR that is bound to DNA.

Physiology and cellular actions of l,25(OH)2D3

In order to delineate the physiologic roles for the vitamin D hormone, it is appropriate first to place the VDR mediator into the context of vitamin D metabolism and cellular actions. Figure 4 summarizes the integration of vitamin D metabolism and cellular actions introduced in Fig. 1, with physiologic regulatory events now super¬ imposed on the metabolic pathway and the inclusion of an expanded list of physiologic actions for the 1,25( )2 4 hormone. The conversion of vitamin D3 to 25(OH)D3 by the liver is a constitutive metabolic step, followed by the 1-hydroxylation of25(OH)D3 to l,25(OH)2D3, a reaction under exquisite control (Haussler & McCain 1977). When blood calcium is low, activation of this latter step occurs, either as a result of the hypocalcémie state per se, or in response to elevated PTH, each of which serves indepen¬ dently to enhance renal 1-OHase activity. Low phosphate is also capable of separately upregulating the 1-OHase enzyme. To limit activation, the hormonal product, l,25(OH)2D3, effects an ultra-short feedback loop to suppress its own biosynthesis in the kidney and also represses PTH synthesis to remove the peptide hormone stimulus of the 1-OHase via a longer feedback loop (Fig. 4). However, the dominant negative feedback controls of 1-OHase activity appear to result from the concerted actions of l,25(OH)2D3 to stimulate bone mineral résorption and to promote intestinal calcium and phosphate absorption, which together elicit an increase in blood calcium and phosphate levels, each of which down-regulates the 1-OHase.

Figure 4 Vitamin D metabolism and cellulat actions, mediated by the VDR-RXR heterodimer binding to a VDRE

The process by which l,25(OH)2D3 causes bone remodeling is complex, involving stimulation of osteoclast differentiation and osteoblastic production of osteopontin, both of which activate résorption in part through the recognition of bone matrix osteopontin by osteoclast surface avß3-integrin. The résorption effect is supported by l,25(OH)2D3-elicited suppression of bone formation via the induction of osteocalcin and the repression of type I collagen. This latter insight that the normal function of osteocalcin is to curtail bone matrix formation arises from the creation of osteocalcin knockout mice (Ducy et al. 1996). In addition to stimulating the transcription of bone-related genes such as osteopontin and osteocalcin, the l,25(OH)2D3 hormone also induces its own eatab¬ olism in kidney as well as other target tissues like bone by enhancing the expression of the vitamin D-24-OHase enzyme. 24-Hydroxylation of l,25(OH)2D3 is the first step in deactivating the hormone, which is eventually metabolized by side chain cleavage to calcitróle acid (Haussler 1986). Thus, the synthesis of l,25(OH),D3 is not only governed by feedback mechanisms that sense l,25(OH)2D3, calcium, PTH and phosphate concentrations, but the hormone induces the termination of its own signal in target tissues, qualifying l,25(OH)2D3 as a bonafide hormone by any definition.

Figure 4 Vitamin D metabolism and cellulat actions, mediated by the VDR-RXR heterodimer binding to a VDRE

As introduced in the section on HVDRR, mediation of the cellular functions of l,25(OH)2D3 requires that VDR bind the hormonal ligand specifically and with high affinity (Fig. 4). Upon such binding, VDR becomes hyperphosphorylated (Jurutka et al. 1993, Haussler et al. 1994) and recruits RXR into a hetero¬ dimeric complex that binds strongly to DNA (Fig. 4). The l,25(OH)2D3-hganded RXR-VDR heterocomplex selectively recognizes VDREs in the promoter regions of positively controlled genes such as osteocalcin (MacDonald et al. 1991), osteopontin (Noda et al. 1990), vitamin D-24-OHase (Ohyama et al. 199A) and ß3-integrin (Cao et al. 1993). Negative VDREs (Haussler et al. 1995) exist in the 5′-regions of the genes for type I collagen (Pavlin et al. 199A), bone sialoprotein (Li & Sodek 1993), PTH (DeMay et al. 1992) and PTH-related peptide (Falzon 1996, Kremer et al. 1996). The mechanisms whereby VDR accomplishes positive and negative control of DNA transcription after VDRE association are not well under¬ stood, although substantial progress has been made in comprehending the stimulation of transcription as detailed in later sections of this article. Moreover, as summarized in Fig. 5, a number of VDREs have been definitively characterized. The prototypical VDBJS is found in the osteocalcin gene, consisting of an imperfect direct repeat of hexanucleotide estrogen responsive element (ERE)-like, half-sites with a spacer of three nucleotides (DR+3). Classic EREs possess a central GT core at positions 3 and 4 of the hexanucleotide, but this feature is only partially conserved in the six natural positive VDREs listed in Fig. 5. There is, however, absolute conservation of the A in position 6 of the 5′ half-element and of the G at position 2 of the 3′ half-element. A preliminary working consensus for the positive VDRE can be derived from these natural VDREs (see boxed sequence in Fig. 5). This generaliz¬ ation is supported, in part, by PCR experiments that were designed to select, from random oligonucleotides, the highest affinity DNA ligand for the RXR-VDR heterodimer (Nishikawa et al. 1994, Colnot et al. 1995).

Figure 5 Natural vitamin D responsive elements (DR+3s) in genes positively tegulated by l,25(OH)2D3. The consensus VDREs are based on either sequence comparisons (boxed) or a selection of random sequences (at bottom).

The random selection process yields an identical VDRE 5′ half-element of GGGTCA (Fig. 5, bottom), which is also a preferred RXR target when RXR homodimers bind to DNA (Yang et al. 1995). This observation is in concert with the conclusion (Jin & Pike 1996) that, with respect to association ofRXR-VDR with VDREs, RXR lies on the 5′ half-element whereas VDR is situated on the 3′ half-element. Examination of both consensus sequences suggests that the G at position 3 of the spacer is important in VDR binding, a deduction consistent with the finding (MacDonald et al. 1991) that this base is partially protected by RXR-VDR in methylation interference assays. How¬ ever, interesting differences arise when one compares the most frequently encountered 3′ half-element bases in natural VDREs, namely the GGGGCA composite which actually occurs in human osteocalcin, with the GGTTCA random consensus selection for the 3′ half-element (Fig. 5). Clearly, GGTTCA represents a potent VDR binding site, a supposition that is bolstered by the fact that osteopontin, which possesses a perfect DR+3 of GGTTCA, is the highest affinity VDRE we have tested (data not shown). Intriguingly, Ts at positions 3 and 4 in the 3′ VDR half-site occur infrequently in the balance of natural VDREs (Fig. 5). The paucity of Ts in the 3′ half-element could be related to a need for varying potency of VDREs in regulated genes, or may even provide for a repertoire of different VDR conformations that could be induced by contact with distinct 3′ half-site core sequences. This postulated range of VDR conforma¬ tions might endow the receptor with the ability to recruit a variety of different coactivators and corepressors, or even to favor the binding of one vitamin D metabolite ligand over another. Irrespective of the above considerations, it is evident that the primary VDRE is a DR+3 recognition site in DNA that directs the VDR to the promoter region of l,25(OH)2D3 regulated genes, ultimately altering the functions of target cells as a result of transcriptional control of gene expression.

Significance of lipophilic ligands in the association of RXR-VDR with DNA

Dimeric complexes are a feature commonly employed in the regulation of eukaryotic transcriptional systems. This process of protein dimerization often will generate novel heterodimeric complexes which display highly cooperative binding to DNA as well as an altered target sequence specificity (Glass 1994). Among the classical steroid hormone receptors, dimerization results in the formation of symmetrical homodimeric protein complexes on palindromic DNA half sites. Dimerization has been shown to be mediated in part by residues within the DNA binding domain of the receptor (Luisi et al. 1991) and is enhanced by residues within the ligand binding domain (Falwell et al. 1990). The other subfamily of nuclear hormone receptors, including VDR, TR and RAR, apparently binds with highest affinity to direct repeat elements either as homodimers or, more commonly, as heterodimers with RXR (Kliewer et al. 1992). In both subgroups of nuclear receptors, protein-protein interactions serve to align the DNA binding domains so that they are optimally positioned to bind to their specific DNA target sequences (Kurokawa et al. 1993, Perlmann et al. 1993, Rastinejad et al. 1995). The ligand binding region of these receptors is multifunctional, in that this domain not only binds the cognate ligand, but also it possesses a dimerization surface as well as the ligand-dependent transactivation function, AF-2 (Gronemeyer 1991, Chambón 1994). The dimerization surface consists of packed helices which are stabilized by hydrophobic heptad repeats interspersed throughout the structure. Ligand apparently can influence different functional components, including the dimerization interface, and the activating AF-2 domain (Renaud et al. 1995, Wagner et al. 1995). Therefore, a likely role for ligand is to regulate the association and dissociation of dimeric protein complexes and hence regulate specific binding to DNA target sequences.

In this regard the following three questions remain regarding l,25(OH)2D3-mediated control of positively regulated genes: (i) does VDR bind as a homodimer (Freedman et al. 1994, Nishikawa et al. 1994) as well as a heterodimer to DR+3 VDREs? (ii) What is the effect of the l,25(OH)2D3 ligand on VDR or VDR-RXR binding to VDREs? (iii) What role does 9-cis retinoic acid, the RXR ligand, play m RXR-VDR binding to VDREs and enhanced transcription of l,25(OH)2D3-responsive genes? It is generally accepted that TR forms homodimers as well as heterodimers with RXR on thyroid hormone responsive elements (TREs), although recent data suggest that the TR homodimer, when unoccupied by thyroid hormone, operates as a repressor of transcription (Chin & Yen 1996, Schulman et al. 1996). Thyroid hormone is proposed to dissociate TR homodimers to facilitate TRRXR heterodimerization on the TRE and stimulate transcription. In contrast, RAR does not appear to be capable of forming homodimers on DR+5 retinoic acid responsive elements (RAREs) (Perlmann et al. 1996), instead cooperating exclusively with RXR in RARE association and vitamin A metabolite-responsive transcrip¬ tion. When present in excess in gel mobility shift DNA binding assays in vitro, both TR and RAR display RXR heterodimeric association with their respective hormone responsive elements (HREs) in the absence of added lipophilic ligand. These in vitro studies are consistent with immunocytochemical data indicating that, unlike classic steroid honnone receptors that reside in the cytoplasm complexed with Hsp-90 and other proteins in their unoccupied state, unliganded TR, RAR and VDR (Clemens et al. 1988) exist in the nucleus in general association with DNA. These findings have led to the dogma that ligand is not required for TR, RAR and VDR to associate with target HREs. Indeed, we have observed that addition of 260 ng baculovirus-expressed hVDR to a gel shift reaction generates weak homodimeric VDR as well as strong VDR-RXR-heterodimeric binding to a rat osteocalcin VDRE probe, both of which are independent of the presence of l,25(OH)2D3 (Nakajima et al. 1994). However, in vivo footprinting experiments (Blanco et al. 1996, Chen et al. 1996) have led to the conclusion that, at least in the case of RAR-RXR heterodimers, RAR ligands are required for RARE binding. We, therefore, sought to devise an in vitro gel shift assay that would more accurately reflect the in vivo situation, primarily consisting of the use of physiologic salt (0-15 m KCl) concentrations and limited amounts of partially purified, baculovirusexpressed VDR and RXRs (Thompson et al. 1997). Utilizing this assay, we have addressed the three questions regarding VDR/RXR listed above, namely heterodimer versus homodimer, the potential role of l,25(OH)2D3 and the effect of 9-cis retinoic acid (9-cis RA).

When 20 ng VDR (~ 10 nM) or 20 ng VDR plus 20 ng RXR are incubated with either the rat osteocalcin or mouse osteopontin VDREs (see Fig. 5), no DNA-bound homodimeric VDR species is apparent, but a VDRE complexed VDR-RXR heterodimer occurs that is strik¬ ingly dependent upon the presence of the l,25(OH)2D3 ligand (Thompson et al. 1997). Thus, at receptor levels approaching that in a typical target cell, a VDR liganddependent heterodimer with RXR is the preferred VDRE binding species. Only when VDR or VDR plus RXR levels are raised to 100 ng of each receptor with the mouse osteopontin VDRE (Thompson et al. 1997), or 260 ng with the weaker rat osteocalcin VDRE (Nakajima et al. 1994), can faint homodimers of VDR bound to the probe be visualized. In addition, at these greater amounts ofreceptors, neither the VDR homodimer nor the VDRRXR heterocomplexes are modulated significantly by inclusion of l,25(OH)2D3 in the incubation (Thompson et al. 1997). We, therefore, conclude that higher receptor levels in vitro generate artifactual VDR homodimers as well as attenuate the normal physiological ligand dependence of VDR-RXR binding to the VDRE. To explain seemingly ligand-independent VDR-RXR association with the VDRE, we postulate the existence of a subpopulation of VDR that is unstably activated in the absence of l,25(OH)2D3 (Schulman et al. 1996) and therefore capable of heterodimerization to generate a positive gel mobility shift under conditions of vast receptor excess. In contrast, our physiologically relevant gel shift assay at <10nM receptor levels and 0-15 m KCl reflects the presumed in vivo events of ligand triggered heterodimerization (Blanco et al. 1996, Chen et al. 1996), and extends earlier in vitro data showing that l,25(OH)2D3 enhances VDRRXR complex formation (Sone et al. 1991, MacDonald et al. 1993, Ohyama et al. 1994).

Next, we tested the effect of 9-cis RA in this gel shift assay. A spectrum of data exists on the role of 9-cis RA in l,25(OH)2D3-stimulated transcription, including demon¬ stration of synergistic action with l,25(OH)2D3 (Carlberg et al. 1993, Schrader et al. 1994, Kato et al. 1995, Sasaki et al. 1995), negligible action (Ferrara et al. 1994), or an inhibitory effect (MacDonald et al. 1993, Jin & Pike 1994, Lemon & Freedman 1996). These marked differences likely result from varying transfection and ligand addition protocols, as well as cell and species specificity. Employing the physiological gel shift procedure with biochemically defined components, we obtained clear evidence that 9-cis RA is a potent inhibitor of l,25(OH)2D3-enhanced, VDR-RXR binding to VDREs such as osteocalcin, with dramatic attenuation by the retinoid occurring at concentrations as low as 10 m (Thompson et al. 1997). Previous gel shift data had also hinted at 9-cis RA inhibition (MacDonald et al. 1993, Cheskis & Freedman 1994), even though higher concentrations of 9-cis RA were utilized in these earlier studies. One somewhat puzzling finding, however, was that the suppressive effect of 9-cis RA seemed more pronounced in vitro than in transfected cells, where retinoid inhibition of l,25(OH)2D3-stimulated transcription is significant, but 50% or less in magnitude (MacDonald et al. 1993). This suggested that multiple pathways may exist for the assembly of the RXR-VDR heterocomplex in vivo. To probe for distinct routes of assembly, we varied the order of addition ofVDR, RXR, l,25(OH)2D3 and 9-cis RA in the gel shift assay for VDRE binding (Thompson et al. 1997). The results showed that 9-cis RA is a potent inhibitor of VDR-RXR heterodimerization on the VDRE in all situations except when VDR alone is preincubated with l,25(OH)2D3 followed by addition of RXR (Thompson et al. 1997). To explain these data, we have developed the model depicted in Fig. 6, which hypothesizes two alternative allosteric pathways for the interaction ofVDR-RXR with the VDRE.

Figure 6 Model of two different allosteric pathways for VDR-RXR-1,25(OH)2D3 binding to DNA.

In pathway A (Fig. 6), l,25(OH)2D3 occupies monomeric VDR, altering the conformation of the ligand binding domain such that it recruits RXR for heterodimeric binding to DNA and subsequent VDRE recognition. Importantly, we pos¬ tulate that previously occupied VDR conformationally influences RXR in the resulting heterodimer such that it is incapable of being liganded by 9-cis RA (pathway A, Fig. 6). This action to abolish RXR ligand responsiveness both silences the ability of 9-cis RA spuriously to trigger vitamin D hormone signal transduction, and prevents 9-cis RA from dissociating the RXR-VDR complex in order to divert RXR for retinoid signal transduction. On the other hand, as illustrated in pathway (Fig. 6), we propose that RXR exists in a different, 9-cis RA-receptive, allosteric state in most other circumstances, such as when present as a monomer, in an apoheterodimer with VDR, or even when the apoheterodimer of RXR and VDR is subsequently liganded with l,25(OH)2D3. This latter species of RXR-VDR-l,25(OH)2D3 (pathway B) is hypothesized to be fully competent in VDRE recognition, but the 9-cis RA binding function of the RXR partner has not been conformationally repressed, rendering this form sensitive to dissociation by 9-cis RA, which would then favor the formation of retinoid-occupied RXR homo¬ dimers. Therefore, unless VDR monomers are first occu¬ pied by l,25(OH)2D3 (pathway A), 9-cis RA can operate to divert or dissociate RXR and direct it to form RXR homodimers (pathway B). It is tempting to speculate that the l,25(OH),D3-liganded heterodimer in pathway A is more potent in transcriptional stimulation than the analogous species in pathway B, perhaps because the AF-2 function of the RXR partner is allosterically activated only in the former instance. The l,25(OH)2D3-occupied VDR-RXR in pathway has the advantage of flexible regulation because it is effectively a two-ligand switch. It likely occurs in vivo because, as stated above, the fact that 9-cis RA blunting significant but incomplete suggests that at least two populations of RXR-VDR heterodimers exist. Finally, when our model (Fig. 6) is compared with those for RXR-RAR and RXR-TR (Forman et al. 1995), it is evident that VDR is closer in mechanism of action to the TR, where 9-cis RA inhibits TR signal transduction by diversion of BJÍR (Lehmann et al. 1993). Also analogous is the fact that thyroid hormone occupation of the TR partner abolishes 9-cis RA binding to the RXR counter¬ part (Forman et al. 1995). Finally, the action of RXRPJ\R heterodimers seems to be fundamentally different from that of RXR-VDR in that RAR liganding by a retinoid facilitates RXR occupation by its retinoid ligand, resulting in cooperative stimulation of gene transcription by the repertoire of vitamin A metabolites.

VDR protein-protein interactions that effect gene transcription

Although we now have at least a rudimentary understand¬ ing of ligand-induced VDR binding to a VDRE, the next logical question is how does VDR regulate the machinery for gene transcription? In the basal state ofDNA transcrip¬ tion, the TATA-box binding protein (TBP) and its associated factors (TAFs) are bound to the TATA box at approximately position — 20 in the 5′ region of controlled genes, but the frequency of transcriptional initiations is very low because the RNA polymerase II-basal transcription factor IIB (TFIIB) enzyme complex is not stably associated with TBP-TAFs. The recruitment of the TFIIB-RNA polymerase II complex appears to be the rate limiting step in preinitiation complex formation, and is stimulated dramatically when a transacting factor or factors bind to upstream enhancers. In a process involving DNA looping, transactivators are thought to attract TFIIB and also interact with TAFs, forming a stable preinitiation complex that executes repeated rounds of productive transcription. Recent data indicate that the activation function in the hormone binding domain of the estrogen receptor, AF-2, associates specifically with a TAF known as TAFn30 (Jacq et al. 1994) and that the estrogen receptor (ER) binds to TFIIB in vitro (lng et al. 1992). In collaboration with Ozato and associates and Tsai and O’Malley, we have observed that hVDR also specifically associates with hTFIIB (Blanco et al. 1995). In this work, Blanco et al. (1995) showed that VDR binds to a TFIIB-glutathione S transferase fusion protein linked to glutathione-laden beads. Additionally, it was observed that both TRa and RARa interact with hTFIIB (Blanco et al. 1995), but that RXR does so only very weakly (P W Jurutka, L S Remus and M R Haussler, unpublished results). This last result suggests that, while the ligand binding partners in the VDR/TR/RAR subfamily provide a hard-wired connection to the assembly and en¬ hancement of the transcription machinery, the RXR partner is not primarily engaged in TFIIB contact.

Independent data obtained by MacDonald et al. (1995) using the powerful yeast two-hybrid system to detect protein-protein interactions also revealed that hVDR binds efficiently to TFIIB. Moreover, MacDonald et al. (1995) further exploited the yeast two-hybrid system to prove that, while hVDR and RXR interact, no homodimeric association occurs for hVDR alone, providing further evidence against the existence of physiologically significant VDR homodimers. Utilizing fusion protein technology, they also showed that VDR interacts directly with RXR to form a heterodimer in solution in the absence of DNA and, further, that this process was enhanced 8-fold by the presence of l,25(OH)2D3 hor¬ mone (MacDonald et al. 1995). Because hVDR-TFIIB association is not dependent upon the l,25(OH)2D, ligand (Blanco et al. 1995, MacDonald et al. 1995), the role of l,25(OH)2D3 can now be further resolved to an early participation in conforming VDR such that it attracts RXR followed by the targeting of the resulting RXR-VDR heterodimer to VDREs (see Fig. 6).

Figure 6 Model of two different allosteric pathways for VDR-RXR-1,25(OH)2D3 binding to DNA.

Interestingly, the presence of BJCR further facilitates VDR-TFIIB association, especially in the presence of l,25(OH)2D3 (PW Jurutka, LS Remus and MR Haussler, unpublished results). In fact, because of its capacity to enhance VDR-RXR heterodimerization, the l,25(OH)2D3 ligand is capable ofgenerating high levels of an RXR-VDR-TFIIB ternary complex in solution, sig¬ nificantly in excess ofthat occurring with either RXR and TFIIB or even with VDR and TFIIB (P W Jurutka, L S Remus and M R Haussler, unpublished results). These data not only reaffirm the interaction ofVDR with TFIIB, but also they imply that the l,25(OH)2D3-liganded VDR-RXR complex is the most efficient binder of TFIIB. This latter effect may be the result of positive conformational influences of RXR on liganded VDR, since VDR is the primary attachment moiety for TFIIB.

Because VDR-TFIIB interactions have been detected either in vitro or in the yeast system where certain mammalian cell restrictions may be relaxed, it was import¬ ant to confirm the relevance ofVDR-TFIIB association in mammalian cells. Blanco et al. (1995) have reported functional studies which, for the first time, show the interaction ofTFIIB with a member ofthe steroid receptor superfamily in ligand-dependent activation oftranscription in intact cells. In pluripotent PI9 mouse embryonal carcinoma cells, transfection of hVDR or hTFIIB alone produced no better than a 2-fold induction of VDREluciferase reporter expression by l,25(OH)2D3. However, when transfected together, hVDR and hTFIIB mediated a synergistic transcriptional response of approximately 30-fold when l,25(OH)2D3 was added, an effect which was absolutely dependent on the presence of the VDRE in the luciferase construct. It should be noted that the VDR-TFIIB positive cooperation appears to be cellspecific because similar experiments in contact-inhibited NIH/3T3 Swiss mouse embryo cells resulted in squelching of transcription by TFIIB. Therefore, in more differentiated cells, perhaps including osteoblasts or fibro¬ blasts, accessory coactivators may be present to modulate TFIIB or bridge between VDR and TFIIB.

In summary, VDR and TFIIB are hypothesized to exist in a multi-subunit transcription complex which also con¬ tains TAFs and/or coactivators that may be promoter- or tissue-specific. Further characterization of this complex will require the discovery of cell type and promoterspecific components via transfection and biochemical interaction studies. Ultimately, an in vitro transcription system must be devised which utilizes defined components to replicate faithfully l,25(OH)2D3-stimulated gene expression.

One subdomain of VDR that likely interacts with coactivators and/or basal transcription factors is the extreme C-terminus. We have previously shown that 403 hVDR, a truncated receptor that lacks the C-terminal 25 amino acids, binds l,25(OH),D3 ligand with reasonable affinity and heterodimerizes normally with RXR, but is devoid of transcriptional activity (Nakajima et al. 1994). These data suggest that VDR contains a transcriptional activation domain near its C-terminus.

Figure 7 The extreme C-terminal amino acid sequence compared across the nuclear receptor superfamily: VDR appears to share the ligand-dependent transcription activation function (AF-2). AR, androgen receptor; CR, glucocorticoid receptor; PR, progesterone receptor.

Indeed, as illustrated in Fig. 7, the region of VDR from residues 416 to 422 possesses a high degree of similarity to the analogous sequences in the entire nuclear receptor superfamily. One hallmark of this conserved sequence is the glutamic acid residue at position 420 of hVDR (Fig. 7) included in a consensus of (where cp=a hydrophobic amino acid) for this domain (Renaud et al. 1995, Wagner et al. 1995). Allowing for conservative replace¬ ments, it seems virtually certain that hVDR forms an amphipathic helix (corresponding to helix 12 in the other receptors) surrounding glutamic acid-420 that is analogous to the ligand-dependent activation function (AF-2) char¬ acterized for TR (Barettino et al. 199A), RAR (Renaud et al. 1995), RXR (Leng et al. 1995) and ER (Danielian et al. 1992). Although this AF-2 domain is capable of autonomously activating transcription (Leng et al. 1995), that such activity is modest may be because of the fact that the AF-2 region is proposed to operate in a liganddependent fashion, involving a structural rearrangement to reposition the AF-2 for both intramolecular and intermolecular protein—protein interactions. Specifically, based upon the crystal structure of unoccupied RXR (Bourguet et al. 1995) and liganded RAR (Renaud et al. 1995) and TR (Wagner et al. 1995), helix 12/AF-2 appears to protrude outward from the more globular ensemble of helices 1—11 in the absence ofligand, such that it is unable to interact efficiently with coactivator/transcription factor. Upon liganding, a conformational signal is then transmit¬ ted to helix 12 that causes it to fold back on helix 11 and attach to the face of the globular ligand binding domain. The pivoting of helix 12 seemingly accomplishes two feats that mediate ligand-activated transcription by the receptor: (i) closing of a ‘door’ on the channel through which the lipophilic ligand enters the internal binding pocket of the receptor, and (ii) locking helix 12 into a stable confor¬ mation that facilitates its interaction with coactivator/ transcription factor. Ligand binding contacts on or near helix 12 (see Fig. 3) probably are significant in maintaining this active positioning of helix 12, essentially trapping ligand in the binding pocket to effect more sustained transactivation events.

Figure 7 The extreme C-terminal amino acid sequence compared across the nuclear receptor superfamily: VDR appears to share the ligand-dependent transcription activation function (AF-2). AR, androgen receptor; CR, glucocorticoid receptor; PR, progesterone receptor.

In order to evaluate the relevance of the above proposed mechanism for VDR action, we (Jurutka et al. 1997) have altered E-420 and L-417 (see Fig. 7) individually to alanine residues, which preserves the putative -helical character of this region. The altered VDRs bind ligand near-normally, with only a mild increase (about 3-fold) in the Kd for the E420A receptor. Both E420A and L417A hVDRs also heterodimerize efficiently with RXR and associate with VDREs similarly to wild-type hVDR, yet their capacity for l,25(OH)2D3-stimulated transcription is abolished, even at high doses ofligand (Jurutka et al. 1997). These point mutations, therefore, identify a C-terminal AF-2 in VDR which corresponds to similar activation domains in other nuclear receptor superfamily members. Because VDR interacts with TFIIB, one of the first questions we asked was whether the VDR AF-2 consti¬ tutes a contact site for this basal transcription factor. Although some very preliminary evidence existed for an association between TFIIB and the C-terminus of hVDR (MacDonald et al. 1995), we observed that neither the E420A nor the L417A mutant VDRs are impaired in their interaction with TFIIB as probed with glutathione-S transferase—TFIIB fusion protein binding technology (Jurutka et al. 1997). Thus, the domain(s) of VDR that interfaces with TFIIB apparently lies elsewhere in the receptor, possibly in the N-terminal portion of the ligand-binding region (Blanco et al. 1995), in the hinge (MacDonald et al. 1995), or in the vicinity of the DNA-binding zinc fingers.

The present experiments with VDR are in concert with recent insight into the function of AF-2 in other nuclear receptors, which is to recruit coactivators of the type of steroid receptor coactivator-1 (SRC-1) (Oñate et al. 1995). A number of candidate coactivators have been isolated in addition to SRC-1 (Halachmi et al. 199A, Baniahmad et al. 1995, CavaiUes et at. 1995, Lee et al. 1995, Hong et al. 1996) and, in several cases, interaction with nuclear receptors requires intact AF-2 core regions (Baniahmad et al. 1995, CavaiUes et al. 1995). Moreover, AF-2 mutations act as dominant negative receptors, for example in the case of hRARy (Renaud et al. 1995). Indeed, we have observed that VDR AF-2 mutants E420A and L417A exhibit dominant negative properties with respect to transcriptional activation (Jurutka et al. 1997). Such AF-2 altered receptors are inactive transcriptionally, but can bind l,25(OH)2D3 ligand and heterodimerize normally on VDREs, the consequence being competition with wild-type VDR-RXR heterodimers for VDRE binding. These data argue that the AF-2 of the primary VDR partner in an RXR-VDR heterodimer is absolutely required for the mediation of l,25(OH)2D3-activated transcription, not only for its intrinsic activation potential, but also because of its presumed role in stabilizing the retention of l,25(OH)2D3 ligand in the VDR binding pocket.

Figure 6 Model of two different allosteric pathways for VDR-RXR-1,25(OH)2D3 binding to DNA.

What part, if any, is played by the AF-2 domain (Fig. 7) of the RXR ‘silent’ partner in the RXR-VDRl,25(OH)2D3 signal transduction pathway? To investigate this phenomenon, AF-2 truncated mutants of RXRa or RXRß were created and tested for their ability to function as dominant negative modulators of l,25(OH)2D3- stimulated transcription (Blanco et al. 1996). Because previous data with RXR-RAR control of gene expression seemed to indicate that the RXR AF-2 was dispensable (Durand et al. 1994), we were surprised to find that AF-2 truncated RXRs were potent dominant negative effectors of l,25(OH)2D3 action in transfected cells (Blanco et al. 1996). We, therefore, conclude that although the RXR ‘silent’ partner in VDR signaling apparently is not occupied by retinoid ligand (see Fig. 6), its AF-2 does play an active role in transcriptional stimulation. A similar conclusion has also been reached recently by two other groups studying RXR-RAR action (Chen et al. 1996, Schulman et al. 1996), with the use of RAR-specific ligands precluding ligand binding by the RXR partner. However, Schulman et al. (1996) have introduced a caveat to the above theory as they point out that AF-2-truncated RXRs in heterodimers become strong, constitutive binders of corepressors like the silencing mediators of retinoid and thyroid hormone receptors (SMRTs). Thus, an alternative explanation to an active coactivator-binding role for RXR AF-2 in heterodimers is that it plays a more passive role in excluding corepressors. In this latter scenario, truncation or point mutation (Schulman et al. 1996) of RXR AF-2 generates spurious corepressor binding rather than compromising coactivator contact. Only additional research into coactivator and corepressor associations of VDR-RXR heterodimers will resolve this issue.

General mechanism for vitamin D hormone action on transcription

In order to provide a working hypothesis for l,25(OH)2D3 action at the molecular level, we have developed the model illustrated in Fig. 8. It is based primarily on data from our laboratory and others studying 1,25(OH)2D3 and VDR, and it relies on the assumed similarities between VDR action and that of TR and RAR. VDR is proposed to exist in target cell nuclei, perhaps very weakly associ¬ ated with DNA, in a monomeric, inactive conformation with the C-terminal AF-2 domain extended away from the hormone binding cavity. Upon liganding with l,25(OH)2D3, VDR assumes an active conformation, with the AF-2 pivoted into correct position for both ligand retention and coactivator contact. In addition, the hormone facilitates interaction of VDR and RXR through a stabilized heterodimerization interface. In turn, 1,25(OH)2D3-occupied VDR may itselffunction as a kind of allosteric regulator of RXR, perhaps by conveying a confonnational signal through the juxtapositioned dimer¬ ization domains to induce the AF-2 ofRXR into an active conformation for coactivator binding. As discussed above (see Fig. 6), the joining of preliganded VDR and unliganded RXR apparently renders the RXR partner unresponsive to binding and either activation or dissocia¬ tion by 9-cis RA. Alternatively, if 9-cis RA encounters RXR monomer first (Fig. 8), or binds to RXR that is complexed with VDR in an apoheterodimer (Fig. 6), the retinoid is able to divert the RXR to generate homo¬ dimers and effectively blunt l,25(OH)2D3-driven transcription In the primary activation pathway pictured in Fig. 8, the RXR-VDR-l,25(OH)2D3 complex recognizes and targets the genes to be controlled through high affinity association with the VDRE in a gene promoter region. Coactivators that are presumed to bind to VDR and RXR AF-2 s are then postulated to link with TAFs/TBP, thereby looping out DNA 5′ of the TATA box. This series of events positions VDR such that it can independently recruit TFIIB to the promoter complex, a process that initiates the assembly of the RNA polymerase II holoenzyme into the preinitiation complex. Precedents exist for transcription factors independently attracting TFIIB, such as hepatocyte nuclear factor-4 (Malik & Karathanasis 1996), as well as for a sequential, two-step pathway for activator-stimulated transcriptional initiation (Struhl 1996, Stargell & Struhl 1996). Using the latter model as an analogy, the VDR activator would contact both TBP/ TAFs (via – coactivator bridges) and TFIIB in order to initiate RNA polymerase II holoenzyme assembly. The order of attachment of these two ‘arms’ of activation has not been determined but, at least, in the case of acidic activators, recruitment to the TATA element precedes interaction with components of the initiation complex (Stargell & Struhl 1996). It is of interest that the mechan¬ ism of l,25(OH)2D3 action depicted in Fig. 8 is not only essential for induction of bone remodeling and other vitamin D functions, but is also self-limiting via 24-OHase induction. In addition, these actions of l,25(OH),D, would be blunted under conditions within a cell where 9-cis RA concentrations dominate over those of l,25(OH)2D3.

Figure 8 Model for transcriptional activation by 1,25(OH)2D3 on the promoter of a target gene

The above described molecular mechanism whereby the vitamin D hormone controls gene expression requires further experimental evaluation. To advance our under¬ standing of the structure/function relationships in VDR, a physical characterization of the structure of VDR via X-ray crystallography will be required. Furthermore, in order to comprehend the genomic action of vitamin D in calcium homeostatic and other target cells, it will be necessary to elucidate the detailed involvement of various RXR isoforms, specific TAFs and novel coactivators/ corepressors that might influence the regulation of differ¬ ent vitamin D-controlled promoters. This information in its entirety should assist in determining the potential role for VDR and l,25(OH)2D3 in the pathophysiology of osteoporosis and other endocrine-related bone diseases.

References

Baniahmad C, Nawaz Z, Baniahmad A, Gleeson MAG, Tsai M-J & O’Malley BW 1995 Enhancement of human estrogen receptor activity by SPT6: a potential coactivator. Molecular Endocrinology 9 34-43.

Barettino D, Ruiz MdMV & Stunnenberg HG 1994 Characterization of the ligand-dependent transactivation domain of thyroid hormone receptor. EMBOJournal 13 3039-3049.

Blanco JCG, Wang I-M, Tsai SY, Tsai MJ, O’Malley BW, Jurutka PW, Haussler MR & Ozato 1995 Transcription factor TFIIB and the vitamin D receptor cooperatively activate ligand-dependent transcription. Proceedings of the National Academy of Sciences of the USA 92 1535-1539.

Blanco JCG, Dey A, Leid M, Minucci S, Park B-K, Jurutka PW, Haussler MR & Ozato 1996 Inhibition of ligand induced promoter occupancy in vivo by a dominant negative RXR. Genes to Cells 1 209-221.

Bourguet W, Ruff M, Chambón , Gronemeyer & Moras D 1995 Crystal structure of the ligand-binding domain of the human nuclear receptor RXR-a. Nature 375 377-382.

Cai Q, Hodgson SF, Kao PC, Lennon VA, Klee GG, Zinmeister AR & Kumar R 1994 Inhibition of renal phosphate transport by a tumor product in a patient with oncogeneic osteomalacia. New England Journal of Medicine 330 1645-1649.

Cao X, Ross FP, Zhang L, MacDonald PN, Chappel J & Teitelbaum SL 1993 Cloning of the promoter for the avian integrin ß3 subunit gene and its regulation by 1,25-dihydroxyvitamin D3. Journal of Biological Chemistry 268 27371-27380.

Carlberg C, Bendik I, Wyss A, Meier E, Sturzenbecker LJ, Grippo JF & Hunziker W 1993 Two nuclear signalling pathways for vitamin D. Nature 361 657-660.

more…

 

The structure of the nuclear hormone receptors.
  • 1Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch at Galveston, 77555-0645, USA.

Steroids. 1999 May;64(5):310-9.   http://www.ncbi.nlm.nih.gov/pubmed/10406480

The functions of the group of proteins known as nuclear receptors will be understood fully only when their working three-dimensional structures are known. These ligand-activated transcription factors belong to the steroid-thyroid-retinoid receptor superfamily, which include the receptors for steroids, thyroid hormone, vitamins A- and D-derived hormones, and certain fatty acids. The majority of family members are homologous proteins for which no ligand has been identified (the orphan receptors). Molecular cloning and structure/function analyses have revealed that the members of the superfamily have a common functional domain structure. This includes a variable N-terminal domain, often important for transactivation of transcription; a well conserved DNA-binding domain, crucial for recognition of specific DNA sequences and protein:protein interactions; and at the C-terminal end, a ligand-binding domain, important for hormone binding, protein: protein interactions, and additional transactivation activity. Although the structure of some independently expressed single domains of a few of these receptors have been solved, no holoreceptor structure or structure of any two domains together is yet available. Thus, the three-dimensional structure of the DNA-binding domains of the glucocorticoid, estrogen, retinoic acid-beta, and retinoid X receptors, and of the ligand-binding domains of the thyroid, retinoic acid-gamma, retinoid X, estrogen, progesterone, and peroxisome proliferator activated-gamma receptors have been solved. The secondary structure of the glucocorticoid receptor N-terminal domain, in particular the taul transcription activation region, has also been studied. The structural studies available not only provide a beginning stereochemical knowledge of these receptors, but also a basis for understanding some of the topological details of the interaction of the receptor complexes with coactivators, corepressors, and other components of the transcriptional machinery. In this review, we summarize and discuss the current information on structures of the steroid-thyroid-retinoid receptors.

 

 Cellular retinoid-binding proteins.
Ong DE1.  Author information
Arch Dermatol. 1987 Dec;123(12):1693-1695a.

A number of specific carrier proteins for members of the vitamin A family have been discovered. Two of these proteins bind all-trans-retinol and are found within cells important in vitamin A metabolism or function. These two proteins have considerable sequence homology and have been named cellular retinol-binding protein (CRBP) and cellular retinol-binding protein, type II (CRBP [II]). A third intracellular protein, cellular retinoic acid-binding protein (CRABP) also is structurally similar but binds only retinoic acid. Although retinol appears to be bound quite similarly by the two retinol-binding proteins, subtle differences are apparent that appear to be related to the different functions of the two proteins. That, coupled with the specific cellular locations of the two proteins, suggests their roles. Cellular retinol-binding protein appears to have several roles, including (1) delivering retinol to specific binding sites within the nucleus and (2) participating in the transepithelial movement of retinol across certain blood-organ barriers. In contrast, CRBP (II) appears to be involved in the intestinal absorption of vitamin A and, in particular, may direct retinol to a specific esterifying enzyme, resulting in the production of fatty acyl esters of retinol that are incorporated into chylomicrons for release to the lymph. Like CRBP, CRABP can deliver its ligand retinoic acid to specific binding sites within the nucleus, sites different from those for retinol. The nuclear binding of retinol and retinoic acid may be part of the mechanism by which vitamin A directs the state of differentiation of epithelial tissue.

 

Interaction of the Retinol/Cellular Retinol-binding Protein Complex with Isolated Nuclei and Nuclear Components
GENE LIAU, DAVID E. ONG, and FRANK CHYTIL
Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
http://jcb.rupress.org/content/91/1/63.full.pdf

Retinol (vitamin A alcohol) is involved in the proper differentiation of epithelia. The mechanism of this involvement is unknown. We have previously reported that purified cellular retinol-binding protein (CRBP) will mediate specific binding of retinol to nuclei isolated from rat liver. We now report that pure CRBP delivers retinol to the specific nuclear binding sites without itself remaining bound. Triton X-100-treated nuclei retain the majority of these binding sites. CRBP is also capable of delivering retinol specifically to isolated chromatin with no apparent loss of binding sites, as compared to whole nuclei . CRBP again does not remain bound after transferring retinol to the chromatin binding sites. When isolated nuclei are incubated with [ 3H]retinol-CRBP, sectioned, and autoradiographed, specifically bound retinol is found distributed throughout the nuclei . Thus, CRBP delivers retinol to the interior of the nucleus, to specific binding sites which are primarily, if not solely, on the chromatin . The binding of retinol to these sites may affect gene expression.

Early histological studies have clearly shown that when animals become vitamin A deficient various epithelial tissues of these animals lose the ability to maintain proper differentiation (1) . However, providing retinol (vitamin A alcohol) to the animal permits tissue repair, with improperly differentiated cells rapidly replaced by normal cells (2) . This indicates that vitamin A has an essential role in cellular differentiation . The action of retinol appears to be mediated by a specific intracellular protein called cellular retinol-binding protein (CRBP). CRBP binds retinol with great avidity and specificity and has been detected in a number oftissues (3, 4) . Recently, CRBP was purified and partially characterized (5, 6) . It is distinct from the well-known serum retinol transport protein called retinol-binding protein (5, 7) . That CRBP plays an important role in the action of vitamin A is suggested by the following observations: It is found complexed with retinol in vivo (4, 8). It binds cis-isomers of retinol with a specificity that parallels the in vivo activity of these isomers (9), Finally, if retinol is first complexed with CRBP, the retinol can bind to the nucleus in a specific and saturable manner (10) . In this study we compare the interaction of the CRBP-retinol complex with isolated nuclei to its interaction with isolated chromatin and follow the fate of both the protein and the ligand . The nuclear binding sites for retinol were localized using autoradiography .

……

The experiments described here were designed to gain insight concerning the still unknown molecular mechanisms by which retinol exerts its effects on the differentiation of epithelia. Alterations in genomic expression appear to be induced in animals fed a retinol-deficient diet, as shown by changes in nuclear RNA synthesis observed in vivo (27-30) as well as in vitro (13) . A working hypothesis has been used that retinol, being a small molecule, might exert its action in a way similar to the accepted model for the mode of action of steroid hormones in differentiation . This model involves binding of the steroid hormone inside the target cell to a specific binding protein called a receptor . The resulting cytoplasmic ligand receptor complex, after undergoing a not fully understood conformational change, translocates to the nucleus . The receptor protein can then be detected in nuclear extracts by its ability to bind specifically the steroid hormone. The receptor steroid complex has been shown to interact with chromatin. Such interaction is believed to lead to an altered expression of the genome, which is the basis for the steroid hormone-induced differentiation (31) .

The steroid hormone model has been used profitably to investigate the mode of retinol action . Indeed, a specific binding protein for retinol, CRBP, was discovered to be present in many tissues (3) . Moreover, after purifying this protein to homogeneity, it was demonstrated that CRBP is able to deliver retinol to the nucleus in a specific manner (10) .

However, we report here a unique feature which appears to be distinct from the steroid hormone model. Using retinol CRBP complex in which the radioactive label is on the protein, we find that CRBP delivers retinol in a specific manner to the nucleus; the retinol associates with chromatin, but the protein itself does not remain bound. This conclusion is based on the observation that the radioactively-labeled protein is still able to deliver retinol inside the nucleus, but it cannot be recovered with the nucleus, in contrast to steroid hormone receptors.

The interaction of the specifically delivered retinol appears to be primarily with chromatin. The outer nuclear envelope is apparently not significantly involved in the interaction as Triton X-100-treated nuclei retain 70% of the retinol binding sites found in intact nuclei. It is still possible that the isolated chromatin and the Triton-treated nuclei contain some of the nuclear matrix and that it is actually the matrix which contains the specific binding sites for retinol. However, preliminary evidence indicates that the specific binding sites remain with a soluble chromatin preparation prepared by mild nuclease digest of nuclei rather than with the nuclear matrix. That the CRBP is necessary for delivering retinol to the nucleus is clearly documented by autoradiography. Free retinol, not bound to CRBP, binds nonspecifically to the nuclei, and to chromatin, and autoradiography shows indiscriminate localization of retinol in the lipid-rich nuclear membrane areas.

The data presented here invite the proposal that the retinolCRBP complex enters the nucleus in some manner which is apparently not dependent on the nuclear membrane. The complex then recognizes a limited number (generally an order of magnitude greater than for steroid hormones) of specific sites on the chromatin where the transfer of retinol from CRBP to these sites takes place. The sites were not detectable and may not be accessible if the retinol is free from CRBP. After the transfer CRBP does not remain associated with the specific sites . The functional significance of the specific interaction between retinol and chromatin remains to be demonstrated .

 

Inhibition of vitamin D receptor-retinoid X receptor-vitamin D response element complex formation by nuclear extracts of vitamin D-resistant New World primate cells.
Most New World primate (NWP) genera evolved to require high circulating levels of steroid hormones and vitamin D. We hypothesized that an intracellular vitamin D binding protein (IDBP), present in both nuclear and cytoplasmic fractions of NWP cells, or another protein(s) may cause or contribute to the steroid hormone-resistant state in NWP by disruption of the receptor dimerization process and/or by interference of receptor complex binding to the consensus response elements present in the enhancer regions of steroid-responsive genes. We employed electromobility shift assay (EMSA) to screen for the presence of proteins capable of binding to the vitamin D response element (VDRE). Nuclear and post-nuclear extracts were prepared from two B-lymphoblastoid cell lines known to be representative of the vitamin D-resistant and wild type phenotypes, respectively. The extracts were compared for their ability to retard the migration of radiolabeled double stranded oligomers representative of the VDREs of the human osteocalcin and the mouse osteopontin gene promoters. A specific, retarded band containing VDR-RXR was identified when wild type cell but not when vitamin D-resistant cell nuclear extract was used in the binding reaction with either probe. In addition, vitamin D-resistant cell nuclear extract contained a protein(s) which was bound specifically to the VDRE and was capable of completely inhibiting VDR-RXR-VDRE complex formation; these effects were not demonstrated with nuclear extract from the wild type cell line or with the post-nuclear extract of the vitamin D-resistant cell line. We conclude that a VDRE-binding protein(s), distinct from IDBP and present in nuclear extract of cells from a prototypical vitamin D-resistant NWP, is capable of inhibiting normal VDR-RXR heterodimer binding to the VDRE.
Reversing Bacteria-Induced Vitamin D Receptor Dysfunction to Treat Chronic Disease: Why Vitamin D Supplementation Can Be Immunosuppressive, Potentially Leading to Pathogen Increase
by J.C. Waterhouse, PhD
Recent attempts to increase vitamin D supplementation to prevent and treat chronic disease have arisen primarily out of observations of low vitamin D levels (25-D) being associated with a variety of diseases. However, new research indicates that these low vitamin D levels are often the result rather than the cause of the disease process, just as in the autoimmune disease, sarcoidosis. Trevor Marshall, PhD, recently summarized this alternative perspective on vitamin D, in a session he co-chaired at the 6th International Congress on Autoimmunity. He and his colleagues presented in silico* and clinical data from the last eight years, indicating that intraphagocytic bacteria are able to block the vitamin D receptor (VDR), and this leads to abnormally low measured vitamin D levels. A second consequence of the bacteria-induced VDR blockage is inhibition of innate immunity. By blocking the VDR, bacteria are able to cause persistent infection and inflammation and thus cause many chronic diseases. Short-term symptom reduction observed from vitamin D supplementation appears to be due to immune suppression by precursor forms of vitamin D that add to the bacterial blockage of the VDR. In silico data also indicates that high levels of vitamin D metabolites suppress antimicrobial peptide production by binding to other nuclear receptors (e.g., thyroid-alpha-1, glucocorticoid). Increasingly, epidemiological, geographical and clinical data are lending support to this model of disease. Studies using more advanced cell culture and molecular techniques are confirming the presence of previously undetected bacteria, including biofilm and cell wall deficient bacteria, as well as “persisters.” A greater understanding of how bacteria resist standard antibiotic approaches is also being gained. A protocol has been developed that is successfully restoring VDR and innate immune function with a VDR agonist and eliminating pathogens with low-dose, pulsed combinations of antibiotics. Immunopathological reactions (a.k.a., Jarisch-Herxheimer reactions) occur due to increased pro-inflammatory cytokines resulting from bacterial killing. The result is an exacerbation of symptoms with each dose of antibiotic, but improvement occurs over the long-term. Remission is being achieved in numerous chronic conditions, including many autoimmune diseases and fibromyalgia, as well as many diseases of aging. Although vitamin D ingestion is avoided as part of this protocol, the evidence indicates that the net result of the protocol is improved vitamin D receptor activation.

Introduction
Vitamin D is a topic of increasing interest and has been implicated in many physiological processes beyond its initially recognized role in calcium absorption and metabolism.1 Vitamin D is found in supplements and a few foods (e.g., fish, liver, egg yolk, fortified products). The majority of vitamin D is produced in the skin when exposed to UV radiation from sunlight. But some have begun advocating consumption of levels of vitamin D above the RDA, and some advocate very high levels, ranging from 1,000 to 5,000 IU or more daily.2 Vitamin D is a secosteroid, with a close resemblance in structure to immunosuppressive steroids. Levels of the various vitamin D metabolites are the result of complex feedback mechanisms involving multiple enzymes and receptors, indicating that it is regulated more like a steroid than a nutrient.1

Short-term symptom reduction has sometimes been observed through increases in sun exposure 3,4 or vitamin D supplementation.5 However, this appears to be due to the anti-inflammatory effect arising from immune suppression, analogous to the effect of a steroid, such as prednisone. If one were to assume that the inflammation is purely pathological, this might be considered beneficial, but evidence that has been accumulating over many decades indicates that inflammation in most chronic diseases is occurring in response to undetected chronic bacterial infection (see below). Since immune suppression can promote the increase of pathogens, the effect of vitamin D supplementation is not likely to be harmless in this situation, but appears to have long-term effects associated with increased levels of bacterial pathogens. The role of this microbiota in producing the inflammation and oxidative stress observed in so many diseases will be discussed near the end of this article.6-8

Vitamin D from food or sun is first converted to 25-D (25-hydroxyvitamin-D) and then converted in a second step to the active 1,25D form (1,25-dihydroxyvitamin-D) that is able to activate the vitamin D receptor (VDR). The type of vitamin D usually measured in the blood is the precursor form, 25-D, rather than 1,25-D, the form that activates the receptor. Activation of the vitamin D receptor is extremely important, as it has numerous effects, including effects on the immune system1 and cancer.9,10 However, recent research indicates that increasing vitamin D via supplementation or sun exposure is not the way to achieve more VDR activation in chronic disease, due to blockage of the VDR by bacterial products.6 This insight has been put to use in a new model of chronic disease and a new protocol.6,8,11-14

A New Perspective on Vitamin D and a New Treatment Approach

Trevor Marshall, PhD, (Murdoch University, Australia) has developed a model of chronic autoimmune and inflammatory diseases in which intraphagocytic bacteria cause disease by producing a substance that binds to and blocks the VDR.1 One such substance has been already identified providing proof of principle.1

  • The VDR is important for adequate innate immune function, including the production of numerous antimicrobial peptides.15

These include

  1. cathelicidin and
  2. beta-defensin,

two of the body’s own arsenal of internally produced antibiotics.

Thus, VDR blockage would seem to be an excellent bacterial strategy, as it would lead to poor innate immune system function and further growth of bacteria and other pathogens. A functioning VDR also appears to be important in controlling cell growth and metastasis, so as to help prevent and control cancerous growths.9,10

A protocol based on this model of disease has been achieving a high rate of improvement/remissions in a wide array of conditions.6,11-14,16-18 It involves the use of

  • a VDR agonist, olmesartan, which is able to activate the VDR effectively and safely.

In addition, low dosages of combinations of select pulsed antibiotics are used to eliminate the bacteria, which also helps restore VDR functioning. The protocol also involves avoidance of vitamin D supplementation. When faced with VDR dysfunction, the evidence indicates that

  • attempting to increase 25-D only adds to the dysregulation of the vitamin D metabolites without being able to adequately overcome the bacteria-induced VDR blockage.6,8

Too much vitamin D can be harmful in two ways, according to Marshall’s work.1,6

  1. In silico data from highly sophisticated molecular modeling shows that high vitamin D levels can block the VDR and thus block innate immune function.18 In addition,
  2. high levels of various vitamin D metabolites can affect thyroid-alpha-1, glucorticoid, and androgen receptors and disrupt hormonal control and further affect innate immune function.1

Thus, any short-term symptom reduction from high levels of vitamin D that may occur is probably occurring at the cost of long-term pathogen increase. This has been supported by observations of patient’s responses over time. In the short-term, even for ten years or more in some cases, the person may feel better with high vitamin D intake. But in the long-term, the chronic infection progresses, because the high 25-D is only adding to the bacterial blockage of the VDR and the suppression of bacterial killing.18

Symptoms increase when the immune system is better able to kill the pathogens, due to the high levels of inflammatory cytokine levels that occur. This is called the immunopathological reaction or Jarisch-Herxheimer reaction.6,11 The symptoms range from pain and fatigue to cognitive impairment and depression, but include numerous other symptoms characteristic of the underlying inflammatory condition.6,11 By suppressing the immune response, vitamin D supplementation may suppress these symptoms in the short-term and may even result in a sort of dependence on vitamin D supplementation or sun exposure to keep the symptoms at bay.

The long-term efficacy of the protocol (sometimes called the Marshall Protocol or MP) in activating the VDR is also supported by improved or stabilized bone density, which is typical in patients on the protocol, if the RDA of calcium is consumed. The protocol replaces vitamin D supplementation with use of the VDR agonist olmesartan (120 to 160 mg in divided doses) and reduces the level of bacteria blocking the VDR with antibiotics and, in this way, is apparently effective in activating the VDR.6,12

Marshall proposes that vitamin D receptor blockage results in the low levels of 25-D that have been observed in numerous diseases. The precursor, 25-D form is the form that is most frequently measured. The VDR blockage typically leads to dysregulation of metabolite levels, and one effect is down-regulation of the conversion of vitamin D to 25-D.1 Thus, according to this perspective, low 25-D levels are the result, not a cause, of the disease process. It follows that a low serum 25-D is not indicative of a true vitamin D deficiency in this situation. Both laboratory19 and clinical findings20 have supported the existence of an apparently similar type of down-regulation of conversion to 25-D.

At the same time that low 25-D is observed, high 1,25-D levels are also usually observed. In fact, elevated 1,25-D has been shown to be a good indicator of inflammatory and autoimmune disease.13,16 When interpreting the results, however, it should be remembered that samples must be frozen until analyzed for accurate 1,25-D results. And occasionally, in cases of quite advanced disease or elderly patients, 1,25-D will be low as well, yet still be consistent with VDR blockage and inflammatory disease.21

Marshall’s protocol was first used to treat sarcoidosis. It is well established that a dysregulation of vitamin D levels, often with very high 1,25-D and low 25-D, occurs in this condition.22 Marshall’s and other’s work has confirmed that this dysregulation also occurs in a wide range of other diseases.12,13,23,24 This pattern of high 1,25-D and low 25-D also exists in VDR knockout mice.25 These mice are genetically engineered to lack a VDR, a situation analogous to a bacteria-blocked VDR.

The very complex relationships among genes, metabolites, enzymes, and receptors that Marshall recently summarized1,6 show that vitamin D is not a mere nutrient. In fact, the active form is a secosteroid transcriptional factor. It is part of a highly regulated and complex system influencing many aspects of metabolism and immune function. There are several feedback and feedforward pathways that influence the levels of various vitamin D forms that Marshall reviewed in depth.1

Marshall was recently invited to co-chair a session on vitamin D at the 6th Annual International Autoimmunity Conference, and he gave one of the keynote presentations of the session.6 Several other presentations were given that support the protocol and model. For example, Perez presented data on treatment response in 20 autoimmune conditions that support Marshall’s model.11 The autoimmune diseases successfully treated in this open-label trial include rheumatoid arthritis, systemic lupus erythematosis, diabetes type 1 and 2, psoriasis, Hashimoto’s thyroiditis, Sjogren’s syndrome, scleroderma, uveitis, myasthenia gravis, and ankylosing spondylitis. Chronic fatigue syndrome and fibromyalgia were shown to respond to the protocol in another presentation.17 And another study indicated that dysregulation of nuclear receptors in the endometrium by vitamin D, along with chronic bacterial infection, can help explain the higher prevalence of some autoimmune diseases in women.26

Epidemiological and Short-Term Clinical and Experimental Data
The in silico and clinical data discussed above provide strong evidence for Marshall’s model, and some might argue it is more reliable than epidemiological and short-term evidence. It is widely recognized that there are many limitations inherent in epidemiological and short-term experimental data due to difficulties in obtaining relevant and accurate results. Confounding factors and the inability to assess the effects of long-term immune suppression from high levels of vitamin D make the results less reliable.13,21 Experiments using animal models have the problem of genetic differences and different disease causation methods.1,13Studies of supplementation are often not randomized and thus are subject to unknown confounding factors that may affect the choice to take vitamin D supplements.13 Furthermore, sun exposure is hard to quantify and is often left out of the analyses. Any of the above can lead to invalid conclusions.

Despite this, a number of recent studies that may be relevant will be discussed here to show that there is much independent support for Marshall’s model among these types of studies. In addition, some lesser-known aspects of some of the studies used to support a high vitamin D intake will be reviewed, which cast doubt on some of their conclusions.

Cancer and All-Cause Mortality
In the case of cancer prevention, a recent randomized controlled trial of calcium and vitamin D by Lappe et al.27 is used to support vitamin D supplementation. However, it has a number of serious limitations. One problem is the assumption that removing the data from the first year is justified. If one looks at Figure 1, in the article by Lappe et al,27 in which the data from the first year was included, there is very little difference between calcium and vitamin D vs. calcium alone throughout the study period. No group of patients was given vitamin D alone. Also, there is not yet long-term data on incidence, since the study lasted only four years. Any reduced incidence may reflect delay in diagnosis. In addition, long-term survival may not ultimately improve. In fact, patients taking vitamin D might even die sooner (see below). In addition to the above critique, a number of published comments have also taken issue with this trial, pointing to other problems and limitations.9,28

Another recent study29 reported finding barely significant lower cancer rates in premenopausal women (95% confidence interval, 0.42-1.0) who consumed more vitamin D. However, they found a marginally significant higher rate of moderately differentiated tumors in postmenopausal women who had higher vitamin D intake. And since postmenopausal women make up a much higher proportion of breast cancer cases, this is particularly concerning. This is just one example of the rather inconclusive, mixed data on vitamin D supplementation that becomes apparent when the vitamin D studies are looked at as a whole (see Discussion section in ref. 29). Even the benefit for premenopausal women is questionable. Bertone-Johnson et al.30 pointed out a quite plausible rationale for the existence of a bias toward low estrogen in those who choose to take vitamin D supplements.

A number of limitations found in the other studies are used as a basis for supporting vitamin D supplementation. For instance, the data is rarely long-term enough and rarely covers all the effects possible. Although there may be an appearance of benefit in the short-term or for subsets of the populations studied, a large, long-term prospective study showed no effect of 25-D on the overall cancer mortality rate in the long-term.31 Freedman et al.31 even showed a suggestion of a negative effect of higher vitamin D levels. There was a non-significant increase in overall mortality in the two groups with 25-D at higher levels (80 to <100 nmol/L: Risk Ratio = 1.21, 95% CI =0.83 to 1.78; =100 nmol/L: Risk Ratio = 1.35; 95% CI = 0.78 to 2.31, where 100 nmol/L corresponds to about 40 ng/ml).

This is in accord with a study in prostate cancer32 (also see discussion in ref. 21) and one in pancreatic cancer33 that found higher cancer rates when 25-D was high. Cancer rates increased among patients with a 25-D level above approximately 32 ng/ml. Evidence regarding solar radiation and geographical/latitudinal analyses are also used as evidence, yet solar radiation has many other effects besides raising 25-D.34,35 Many other relevant factors, such as pathogen distributions, climate effects on pathogen spread36,37 and host susceptibility,38 diet, and pollution levels also vary with geographical location.

It was recently pointed out in the Bulletin of the World Health Organization that high 25-D has been found to be associated with greater cancer risk in some studies.39 Studies mentioned, included one that found that there was a higher rate of many internal cancers in patients who have a type of skin cancer that is considered to be the best indicator of long-term sun exposure.40 Another study discussed failed to find a geographical pattern that would support a protective effect of increased 25-D.41 On the whole, in these epidemiological studies, the data is mixed and inconsistent, which is to be expected when there are so many unknown confounding factors affecting 25-D levels and disease incidence that may bias the results.13 In addition, a recent large prospective study presented evidence suggesting that circulating 25-D concentrations may be associated with increased risk of aggressive prostate cancer.42 For all types of prostate cancer, the data failed to support the hypothesis that higher vitamin D decreases prostate cancer risk.42

Studies looking at overall mortality benefits of vitamin D are sometimes misleading at first glance. In the large meta-analysis done recently on the effect of vitamin D and calcium on mortality rates,43 the abstract attributes reduced mortality to vitamin D, yet the only statistically significant results were for calcium together with vitamin D. Another serious problem is that most of the studies analyzed in the meta-analysis were only a few years in duration, so long-term effects on mortality and morbidity could not be accurately assessed.

Bone Density, Parathyroid Hormone
Another area that should be re-evaluated is the negative association between parathyroid hormone and 25-D levels. This association is often used to assert that high levels of 25-D (e.g., 40 –50 ng/ml or more) are optimal. Aloia et al.44 has pointed out that the studies that conclude these high levels of vitamin D are needed fail to require adequate calcium intake, and that is why such high levels are suggested. It should also be considered whether both low 25-D and high PTH are due to the disease process rather than the low 25-D causing the elevated PTH. In addition, only a small percentage of patients with low 25-D have elevated PTH. The low 25-D may be indicating a systemic chronic bacterial infection, and the abnormally high PTH levels in a small percentage of patients may merely be pointing to those cases in which bacteria have infected the parathyroid gland to a greater degree.

In a study comparing vitamin D supplementation with calcium supplementation,45“the effect of calcium on bone loss was blunted in subjects with the highest levels of serum 25OH vitamin D [25-D].” This last finding is supportive of Marshall’s in silico work indicating that high 25-D actually blocks the VDR.6,18 The largest meta-analysis so far clearly showed benefit from calcium supplementation; however, benefit for vitamin D was much less clear.46 No significant benefit for fracture risk was found when comparing vitamin D and calcium to calcium alone, though some differences were found between vitamin D levels.

Another factor that needs to be considered is whether immune suppression is the cause of bone density improvement when high vitamin D levels are used. Immunosuppressive drugs that lower TNF-alpha using antibodies can improve bone density by reducing inflammation.47 High levels of vitamin D supplementation can also lower TNF-alpha48 and suppress the immune response. Thus, it is possible that an increase in bone density from vitamin D supplementation could be the result of immune suppression via TNF reduction, rather than correction of a vitamin D deficiency. TNF-lowering drugs such as infliximab (Remicade) increase risk of cancer and tuberculosis. Thus, the desirability of improving bone density through immune suppression is questionable. This immunosuppressive effect of vitamin D may even explain what seems to be a beneficial effect on falls and muscle strength of elevating vitamin D through supplementation.21 This may be only a symptom reduction in the short-term and may be harmful in the long-term due to the immune suppression.

Autoimmune Disease
In the area of autoimmune disease, the data is equally mixed, and sometimes the larger, more recent studies fail to show any effect of vitamin D levels. For example, a recent large study failed to find an association between serum 25-D levels and the incidence of systemic lupus erythematosis and rheumatoid arthritis.49 Research has found that the average age at which patients acquired rheumatoid arthritis is 12 years earlier in Mexico than in Canada and pointed to the possible role of infectious agents in causing the disease.50 And clearly this study does not support the idea that sun exposure is beneficial for rheumatoid arthritis, since Mexico gets far more sun than Canada.

Although some studies in type 2 diabetes have indicated vitamin D supplementation may be preventive,51 these studies were not randomized and thus are subject to many known and unknown confounding factors affecting a parent’s decision to give a child supplemental vitamin D.13 And even if it were clearly established that vitamin D supplementation reduced the incidence of diabetes in infants and small children, that would not mean that it would help in established disease or older patients, nor would it necessarily mean it is the optimal way to achieve diabetes prevention and long-term health. The positive response of both type 1 and type 2 diabetes patients to the Marshall Protocol11 indicates research on the role of bacteria in diabetes should be a priority.

Influenza and Colds
It has been proposed that vitamin D levels’ decline in winter best accounts for the seasonality of colds and influenza52 and that this potentially supports the need for increased supplementation.52,53 However, new evidence indicates that changes in the viral coat properties can account for the seasonal outbreaks at higher latitudes.36,37 Effects on the airways in dry, cold climates also appear to increase susceptibility to viral and bacterial infections in winter and could contribute to higher winter prevalence of respiratory infections in cold climates.38

Another important point is that the patients being followed on the Marshall Protocol include a number of individuals who report that during the worst period of their chronic illness, they had few or no colds or flu-like illnesses, sometimes for many years at a time. And sometimes this low rate of colds was apparent even years before their illness. This has also been reported in Parkinson’s disease, with the decrease in viral respiratory infections also occurring several years before the disease was diagnosed.54 Thus, even if future research were to establish that vitamin D supplementation reduced colds and influenza, this is by no means an adequate argument for its use. The above observations in chronically ill patients indicate that observing a reduction in respiratory viral infections is not always a sign of good overall health.

Indications of Long-Term Negative Effects of Vitamin D Supplementation
Brannon et al.55 pointed out in a recent report from a roundtable discussion of vitamin D data needs that many studies so far have not yet adequately investigated potential negative consequences such as soft tissue calcification. Vitamin D has been implicated in arterial calcification in the past56 as well as other negative effects.13 The report by the roundtable of vitamin D experts expressed concern that many studies may be shortsighted with regard to adverse outcomes.55

A disturbing new study showed a highly significant correlation (p=0.007) between increased vitamin D intake from food and supplements and the volume of brain lesions shown by MRI in elderly adults.57 In the multivariable regression model, vitamin D intake retained its significant correlation with brain lesion volume even after the effects of calcium were statistically removed. However, calcium did not retain a significant independent correlation with the lesions when the study controlled for vitamin D. Thus, the analysis points to vitamin D supplementation as the key factor in higher lesion volume in this study. These types of brain lesions have been linked to adverse effects in many studies, e.g., stroke,58 psychiatric disorders,59,60 brain atrophy,61 and earlier death.62 Interestingly, the levels of vitamin D intake were not particularly high by some standards, with the highest intake estimated at 1015 mg daily (mean of 341 mg), about half coming from supplements and the rest from food.

The correlation between vitamin D intake and brain lesions seems to lend further support to Marshall’s work. In another study, the finding that over a three-year period, a small percentage of patients were found to have a slight regression of their brain lesions,63 leaves room for hope that the lesions are potentially reversible. Reversibility would be in accord with the improvement of depression and cognitive deficits and other neurological symptoms reported in patients on the Marshall Protocol.6,64

Elusive Bacterial Pathogens Are Detected with Improved Methods
Over many decades, researchers have reported evidence that hard-to-detect bacterial infections are the cause of many diseases,65,66 including autoimmune disease,65-68 cardiovascular disease,69-71 and even cancer.72-77 Some have noted the recent trend toward finding more infectious causes of disease and suggested this is likely to increase in the coming years.6,71,77-80

Recently, Barry Marshall received the Nobel Prize for discovering that the bacteria Helicobacter pylori causes ulcers. And it is now known that H. pylori is a causal factor in stomach cancer.77

New techniques using 16s ribosomal RNA shotgun sequencing,81,82 as well as more advanced culturing and observational techniques65,66,80,83-85 are suggesting that, up until now, most microbiologists have failed to detect a large percentage of potential disease-causing agents. “Persister” cells have been identified that escape antibiotic treatment.86 Cell wall deficient organisms have long been studied,65-66and just recently, advances have been made in understanding their structure and in culturing techniques.80 Research is also indicating that a bacterial biofilm-like microbiota of multiple species even exists within human cells.6,8

Bacteria that grow on a surface in a multi-species community, protected by both a biofilm and the combined effect of their individual resistance strategies, have been a growing area of research.79 Bacterial biofilms have been found to cause the non-healing ulcers in diabetics and may be successfully treated using novel approaches, thus reducing the need for limb amputation.88

Other examples of studies detecting unexpected bacterial pathogens include work linking pathogens in amniotic fluid to pre-term birth89 and research showing numerous previously undetected species in the biofilms that coat prosthetic hip joints.82 Many species of bacteria have been in wounds that were previously undetected using older techniques.81 Macfarlane et al.90 used a combination of more advanced techniques to study bacteria in biofilm communities in patients with Barrett’s esophagus, a pre-cancerous condition. Their methods revealed significant differences between patients and controls in the types and numbers of bacterial species, differences that were previously undetected using older techniques.

Increasingly, inflammation is observed in chronic diseases ranging from depression to cardiovascular disease and cancer.87 The above trends, when combined with observations of bacteria in numerous diseases6,13,65,66,71,91 and the success of the anti-bacterial protocol developed by Marshall6,8,11,13 suggest an extensive role for previously unidentified chronic bacterial infections.

Research is also supporting the ineffectiveness of most standard antibiotic protocols against these bacteria70 and suggesting why other approaches may work better. For instance, some antibiotics target cell walls, and this actually promotes the production of cell wall deficient forms of bacteria that resist many antibiotics.80 Furthermore, many antibiotics are known to inhibit phagocytosis and other aspects of the immune response when taken at high, constant dosages.92

The ability of bacteriostatic antibiotics such as clindamycin to be effective at low doses has been documented.93,94 The survival of “persister” cells mean that pulsed antibiotics are likely to be more effective.86 And fascinating investigations of biofilm communities have revealed many ways in which bacteria can resist antibiotics when used in traditional ways.95 The existence of communities of many bacterial species means that combinations of antibiotics are probably needed to be effective against all the species present. Thus, there is increasing support for the use of pulsed, low dosages of combinations of bacteriostatic antibiotics as used in the anti-bacterial protocol discussed here.

What is particularly encouraging is that the effectiveness of Marshall’s protocol in many systemic chronic disease indicates that these elusive pathogens do respond to select currently available bacteriostatic antibiotics when innate immune function is restored through restoring vitamin D receptor function.6,11 Not only do the bacterial infections appear to resolve, the evidence so far suggests that the improved immune response leads to reduced viral, fungal, and protozoal infections as well.

Conclusions
In silico and clinical data indicate that it is likely that associations between low vitamin D levels and chronic diseases are not evidence of deficiency, but result from a bacteria-induced blockage of the vitamin D receptor, leading to down-regulation of 25-D levels.1,6 According to this model of chronic disease, the short-term benefits sometimes perceived with high vitamin D levels are not due to correction of a vitamin D deficiency but due to suppression of bacterial killing and the immunopathological reaction that accompanies it. Data on reversal of a range of inflammatory and autoimmune diseases through an anti-bacterial protocol that includes vitamin D avoidance and a VDR agonist support this view.6,11

As discussed in detail above, it appears that increasing vitamin D supplementation is not the answer to these chronic diseases and is likely to be counter-productive. Other researchers have also raised concerns regarding vitamin D supplementation’s potential adverse effects. Potential dangers include increased aortic calcification55,56 and brain lesions shown by MRI57 (also see above). In addition, some studies have even found evidence of increased danger from cancer in association with higher levels of vitamin D.32,33,39,40,42

Many have been attracted to the area of vitamin D research, recognizing interesting patterns and responses to supplementation that at first seemed to indicate widespread deficiency and, at the very least, indicate that vitamin D plays a powerful role in physiological processes. Great strides have been made in the last 30 years by scientists with a range of perspectives, and this has led to great excitement and a laudable commitment to use that knowledge to help patients.

However, new genomic and molecular research and the positive response to a new anti-bacterial protocol that involves the avoidance of vitamin D indicate the need for a reappraisal of the data gathered so far. It appears that attempting to raise 25-D through vitamin D supplementation or sun exposure is not the right approach to many, if not most, common chronic diseases. Instead, as discussed above, the evidence supports the effectiveness of a new protocol in restoring vitamin D receptor function, which appears to be a crucial factor in recovery.

One of the most commendable attributes of a truly objective scientist is the willingness to be open to changing long-held positions in the light of new evidence. It will be interesting to see how many have this all-too-rare quality, as research and discussion of vitamin D and the VDR continues. It is to be hoped that the tremendous healing potential likely to be available from eliminating the pathogens that cause chronic disease will inspire an especially high level of open-minded discussion and cooperation.

Caution: The immunopathological reactions from killing the high levels of bacteria that have accumulated in chronically ill patients can be severe and even life-threatening, and thus the Marshall Protocol must be done very carefully and slowly, according to the guidelines.7,96 For the sake of safety, antibiotics must be started at quite low dosages, starting with only one antibiotic. Health care providers are responsible for the use of this information. Neither Autoimmunity Research, Inc., nor the author assume responsibility for the use or misuse of this protocol.

Note: Neither the author, Prof. Marshall, nor the non-profit Autoimmunity Research, Inc. have any financial connection with any product or lab mentioned with regard to the Marshall Protocol. The information needed to implement the Marshall Protocol is available free of charge fromwww.AutoimmunityResearch.org.

Vitamin D3 and Its Nuclear Receptor Increase the Expression and Activity of the Human Proton-Coupled Folate Transporter

Folates are essential for nucleic acid synthesis and are particularly required in rapidly proliferating tissues, such as intestinal epithelium and hemopoietic cells. Availability of dietary folates is determined by their absorption across the intestinal epithelium, mediated by the proton-coupled folate transporter (PCFT) at the apical enterocyte membranes. Whereas transport properties of PCFT are well characterized, regulation of PCFT gene expression remains less elucidated. We have studied the mechanisms that regulate PCFT promoter activity and expression in intestine-derived cells. PCFT mRNA levels are increased in Caco-2 cells treated with 1,25-dihydroxyvitamin D3 (vitamin D3) in a dose-dependent fashion, and the duodenal rat Pcft mRNA expression is induced by vitamin D3 ex vivo. The PCFTpromoter region is transactivated by the vitamin D receptor (VDR) and its heterodimeric partner retinoid X receptor-α (RXRα) in the presence of vitamin D3. In silico analyses predicted a VDR response element (VDRE) in the PCFT promoter region −1694/−1680. DNA binding assays showed direct and specific binding of the VDR:RXRα heterodimer to the PCFT(−1694/−1680), and chromatin immunoprecipitations verified that this interaction occurs within living cells. Mutational promoter analyses confirmed that the PCFT(−1694/−1680) motif mediates a transcriptional response to vitamin D3. In functional support of this regulatory mechanism, treatment with vitamin D3 significantly increased the uptake of [3H]folic acid into Caco-2 cells at pH 5.5. In conclusion, vitamin D3 and VDR increase intestinal PCFT expression, resulting in enhanced cellular folate uptake. Pharmacological treatment of patients with vitamin D3 may have the added therapeutic benefit of enhancing the intestinal absorption of folates.

Folates are water-soluble B vitamins that act as one-carbon donors required for purine biosynthesis and for cellular methylation reactions. They are essential for de novo synthesis of nucleic acids, and thus for production and maintenance of new cells, particularly in rapidly dividing tissues such as bone marrow and intestinal epithelium (Kamen, 1997). Adequate dietary folate availability is especially important during periods of rapid cell division, such as during pregnancy and infancy. Folate deficiency has been associated with reduced erythropoiesis, which can lead to megaloblastic anemia in both children and adults (Ifergan and Assaraf, 2008). Deficiency of folate availability in pregnant women has been linked to neural tube defects, such as spina bifida, in children (Pitkin, 2007). This has prompted the application of folate supplementation schemes either as pills or via fortification of grain products with folates (Eichholzer et al., 2006). Folates have also been proposed to act as protective agents against colorectal neoplasia, although contradictory results have also been reported (Sanderson et al., 2007).

The availability of diet-derived folates is primarily determined by the rate of their uptake into the epithelial cells of the intestine, mediated by the proton-coupled folate transporter (PCFT, gene symbol SLC46A1), localized at the apical brush-border membranes of enterocytes (Subramanian et al., 2008a). PCFT is an electrogenic transporter that functions optimally at a low pH (Qiu et al., 2006;Umapathy et al., 2007). Despite being abundantly expressed in enterocytes, the second folate transporter, termed reduced folate carrier (RFC, gene symbolSLC19A1), has recently been shown not to play an important role in intestinal folate absorption (Zhao et al., 2004; Wang et al., 2005).

The human PCFT gene resides on chromosome 17, contains 5 exons, and is expressed as two prominent mRNA isoforms of 2.1 and 2.7 kilobase pairs (Qiu et al., 2006). Mutations in the PCFT gene have been associated with hereditary folate malabsorption, a rare autosomal recessive disorder (Qiu et al., 2006; Zhao et al., 2007). The PCFT protein is predicted to have a structure harboring 12 transmembrane domains (Qiu et al., 2007; Subramanian et al., 2008a). Although the transport function of PCFT has been studied extensively, relatively little is known about the regulation of PCFT gene expression. PCFT promoter activity has been shown possibly to be epigenetically regulated by its methylation status in human tumor cell lines (Gonen et al., 2008). Furthermore, both the PCFT mRNA expression levels and PCFT promoter activity positively correlate with the level of differentiation of colon-derived Caco-2 cells (Subramanian et al., 2008b).

In addition to its well known roles in regulating calcium homeostasis and bone mineralization, 1,25-dihydroxyvitamin D3 (vitamin D3), the biologically active metabolite of vitamin D, executes many other important functions, particularly in the intestine. For example, vitamin D3 promotes the integrity of mucosal tight junctions (Kong et al., 2008). Many effects of vitamin D3 are mediated via its action as a ligand for the vitamin D receptor (VDR; gene symbol NR1I1), a member of the nuclear receptor family of transcription factors (Dusso et al., 2005). VDR typically regulates gene expression by directly interacting with so-called direct repeat-3 (DR-3; a direct repeat of AGGTCA-like hexamers separated by three nucleotides) motifs within the target promoters, as a heterodimer with another nuclear receptor, retinoid X receptor-α (RXRα; gene symbol NR2B1) (Haussler et al., 1997). Genetic variants of VDR have been associated with inflammatory bowel disease (Simmons et al., 2000; Naderi et al., 2008). Similarly to folates, both VDR and its ligand vitamin D3 have been proposed to be protective against intestinal neoplasia (Ali and Vaidya, 2007). Dietary folate intake has been suggested to regulate gene expression of the components of the vitamin D system, possibly via epigenetic control through the function of folates as methyl donors (Cross et al., 2006). Several intestinally expressed transporter genes, such as those encoding the multidrug resistance protein 1 and multidrug resistance-associated protein 2, have recently been shown to be induced by vitamin D3 (Fan et al., 2009). We investigated whether vitamin D3 regulates the expression of the PCFT gene, encoding a transporter crucial for intestinal folate absorption. The human well polarized enterocyte-derived Caco-2 cells exhibit many of the characteristics associated with mature enterocytes and were used here to investigate the effects of vitamin D3 on PCFT gene expression and folate transport activity.

……..

Vitamin D3 regulates the expression of its target genes primarily by acting as an agonistic ligand for its DNA-binding nuclear receptor VDR, although nongenomic actions by vitamin D3 have also been described previously (Christakos et al., 2003;Dusso et al., 2005). VDR, an important regulator of differentiation and proliferation of enterocytes, typically activates gene expression by heterodimerizing with its nuclear receptor partner RXRα. VDR:RXRα heterodimers then directly bind to DR-3-like elements on the target genes. It should be noted that other modes of VDR-mediated regulation, either via direct interaction with other DNA-binding factors or through nongenomic actions, have also been reported (Dusso et al., 2005).

Here we demonstrate that VDR is a ligand-dependent transactivator of the humanPCFT gene, coding for a vital transporter for intestinal absorption of dietary folates. PCFT mRNA is also abundantly expressed in the liver (Qiu et al., 2006). However, VDR is expressed at very low levels in primary human hepatocytes or hepatocyte-derived cell lines (Gascon-Barre et al., 2003; data not shown), suggesting that VDR-mediated regulation of the PCFT gene may not occur in hepatocytes.

Endogenous PCFT mRNA levels were induced by vitamin D3 in a dose-dependent manner in Caco-2 cells (Fig. 1A). This increase was not further enhanced by cotreatment of cells with the RXRα ligand 9-cis retinoic acid (data not shown), consistent with a previous report that VDR:RXRα heterodimers, at least in some promoter contexts, may not respond to RXRα ligands (Forman et al., 1995). Alternatively, saturating levels of RXRα ligands may already be endogenously present in cells in these experimental conditions. In transient transfection assays, the PCFT promoter fragment −2231/+96 exhibited significant response to exogenous expression of VDR alone in the presence of its ligand (Fig. 2), most probably supported by endogenously expressed RXRα in Caco-2 cells.

Supporting the importance of the VDR:RXRα heterodimer formation for PCFTpromoter regulation, the luciferase values were further significantly elevated upon exogenous expression of RXRα. Exogenous expression of VDR in the absence of vitamin D3 did not notably influence the activity of the PCFT(−2231/+96) promoter, indicating ligand-dependence of VDR action. In deletional transfection analysis, the strongest induction in response to VDR and RXRα in the presence of their ligands was achieved with the PCFT(−2231/+96) promoter fragment (Fig. 3A). Induction of the shortest deletion variant tested [PCFT(−843/+96)luc] was approximately 50% of that achieved for the PCFT(−2231/+96), indicating that this more proximal region is likely to contain further DNA elements mediating a response to vitamin D3. However, in our current study, we focused on the distal region between the nucleotides −2231 and −1674 upstream of the transcriptional start site of the human PCFT gene, which confers maximal response to vitamin D3. In our computational analysis, we identified a putative VDRE within the PCFTpromoter region between nucleotides −1694 and −1680. We have not so far been successful in identifying further binding sites for the VDR:RXRα heterodimer in the more proximal region of the PCFT promoter. It may be that, in addition to direct DNA-binding to the PCFT(−1694/−1680) element identified here, VDR may also affect PCFT promoter activity indirectly, via interactions with other DNA-binding factors. For example, it has been proposed that the p27Kip1 gene is regulated by VDR via response elements for unrelated DNA-binding transcription factors Sp1 and NF-Y (Huang et al., 2004).

Both endogenously expressed and recombinant VDR and RXRα bound to thePCFT(−1694/−1680) element specifically and as obligate heterodimers (Fig. 4). The interaction between VDR and this region of the PCFT promoter within living cells treated with VDR and RXRα ligands was confirmed by chromatin immunoprecipitation tests (Fig. 5). Heterologous promoter assays proved that thePCFT(−1694/−1680) element can function as an independent VDR response element. The significant decrease in VDR:RXRα-mediated induction upon mutagenesis of the PCFT(−1694/−1680) element confirmed that it is an important functional mediator of the effect (Fig. 6, A and B).

Although we observed vitamin D3-mediated increase of rat Pcft mRNA expression ex vivo (Fig. 1C), the rat Pcft promoter (chromosome 10; GenBank accession number NW_047336) exhibits no significant overall homology with the humanPCFT promoter over the proximal 3000-bp regions. This suggests that despite the divergence of the promoter sequences between human and rodent PCFT/Pcftgenes, the functional response to vitamin D3 is conserved.

The activation of PCFT gene transcription by VDR also translates into an increase in PCFT protein function. Vitamin D3 treatment of Caco-2 cells led to significantly increased uptake of folate across the apical membrane, in a dose-dependent manner (Fig. 7). In keeping with the fact that PCFT strongly prefers an acidic milieu for its transport function (Qiu et al., 2006; Nakai et al., 2007; Unal et al., 2009), we only observed vitamin D3-stimulated transport activity at pH5.5, but not at neutral pH. These data strongly suggest that vitamin D3-mediated transcriptional activation of PCFT gene expression leads to an increase of PCFT transport function. Consistent with our model, mRNA expression of the other known folate carrier expressed in Caco-2 cells, RFC, which functions efficiently at neutral pH (Ganapathy et al., 2004; Wang et al., 2004), was not affected by vitamin D3treatment (Fig. 1B). It has been reported that vitamin D3-induced gene expression increases as Caco-2 cells differentiate (Cui et al., 2009). Thus, our current findings on VDR-mediated regulation of PCFT expression provide a possible molecular mechanism for a prior observation that folate uptake into Caco-2 cells is enhanced upon confluence-associated differentiation (Subramanian et al., 2008b).

Our results suggest that intestinal folate absorption may be enhanced by an increase in dietary vitamin D3 intake. Food products are often supplemented with folates, because of their proposed beneficial health effects. Based on our current study, supplementation of vitamin D3 may enhance the intestinal absorption of folates. PCFT also transports the antifolate drug methotrexate (MTX) (Inoue et al., 2008; Yuasa et al., 2009) widely used in the treatment of autoimmune diseases and cancer. MTX interferes with folate metabolism by competitively inhibiting the enzyme dihydrofolate reductase. Our results may further suggest a potential mechanism to increase intestinal absorption of MTX via simultaneous treatment with vitamin D3, thereby affecting the bioavailability of MTX. Patients suffering from inflammatory bowel disease are frequently on long-term treatment with calcium and vitamin D3 as a prophylaxis against osteopenia and osteoporosis (Lichtenstein et al., 2006). This patient group is frequently treated with folates (in the case of folate deficiency) or MTX (as a second-line immunosuppressant) (Rizzello et al., 2002). MTX therapy per se requires prophylactic administration of folates, and these patients often receive additional calcium/vitamin D3. Our current results may warrant a closer investigation into potential drug-drug interactions between pharmacologically administered vitamin D3, MTX, and folates. Taking into account the previous report that folates regulate the expression of genes involved in vitamin D3 metabolism, it may be that folate and vitamin D3 homeostasis are closely interlinked through such mutual regulatory interactions.

 

Innate immune response and Th1 inflammation
http://mpkb.org/home/pathogenesis/innate_immunity

The innate immune response is the body’s first line of defense against and non-specific way for responding to bacterial pathogens.1 Located in the nucleus of a variety of cells, the Vitamin D nuclear receptor (VDR) plays a crucial, often under-appreciated, role in the innate immune response.

When functioning properly, the VDR transcribes between hundreds2 and thousands of genes3including those for the proteins known as the antimicrobial peptides. Antimicrobial peptides are “the body’s natural antibiotics,” crucial for both prevention and clearance of infection.4The VDR also expresses the TLR2 receptor, which is expressed on the surface of certain cells and recognizes foreign substances.

The body controls activity of the VDR through regulation of the vitamin D metabolites. 25-hydroxyvitamin D (25-D) antagonizes or inactivates the Receptor while 1,25-dihydroxyvitamin D (1,25-D) agonizes or activates the Receptor.

Greater than 36 types of tissue have been identified as having a Vitamin D Receptor.5

Another component of the innate immune response is the release of inflammatory cytokines. The result is what medicine calls inflammation, which generally leads to an increase in symptoms.

Before the Human Microbiome Project, scientists couldn’t link bacteria to inflammatory diseases. But with the advent of DNA sequencing technology, scientists have detected many of the bacteria capable of generating an inflammatory response. All diseases of unknown etiology are inflammatory diseases.

Nuclear receptors and ligands

Nuclear receptors are a class of proteins found within the interior of cells that are responsible for sensing the presence of hormones and certain other molecules. A unique property of nuclear receptors which differentiate them from other classes of receptors is their ability to directly interact with and control the expression of genomic DNA. Some of the molecules (or ligands) which bind the nuclear receptor activate (agonize) it and some inactivate (antagonize) it.

It is commonly accepted that most ligands, approximately 95% to 98%, inactivate the nuclear receptors. Since the nuclear receptors play a significant role in the immune response, this factor alone may explain why so many drugs and substances found in food and drink are immunosuppressive.

Because the expression of a large number of genes is regulated by nuclear receptors, ligands that activate these receptors can have profound effects on the organism. Many of these regulated genes are associated with various diseases which explains why the molecular targets of approximately 13% of FDA approved drugs are nuclear receptors.6

Different cell types have different nuclear receptors. One of the nuclear receptors seen in immune cells is the Vitamin D Receptor (VDR). The VDR has two endogenous or “native” ligands, which are also the two main forms of vitamin D in the human body: 25-hydroxyvitamin D (25-D) and 1,25-dihydroxyvitamin D (1,25-D). Non-native or exogenous ligands can also inactivate or activate a nuclear receptor, depending on its molecular structure.

Ligands compete to dock at nuclear receptors. When is a given kind of ligand such as 25-D as opposed to 1,25-D more likely to bind to the VDR? It depends. 1,25-D tends to be much less common than 25-D – by a factor of 1,000 or more – so it binds to the receptor much more infrequently. A greater concentration of a given molecule can displace competing molecules off the nuclear receptor. Affinity occurs in logarithmic fashion, which is to say that it operates on the basis of a sliding scale. In short, an increase in 1,25-D and a decrease in 25-D can tilt the odds in favor of 1,25-D, and vise versa.

Affinity as well as the question of whether a ligand inactivates or activates a nuclear receptor can all be validated using in silicomodeling. Although less precise, it is also possible to measure these properties in vitro.

Activated by 1,25-D and inactivated by 25-D, the Vitamin D nuclear receptor (VDR) transcribes a number of genes crucial to the function of the innate immune response.

Role of Vitamin D Receptor in innate immunity

Vitamin D/VDR have multiple critical functions in regulating the response to intestinal homeostasis, tight junctions, pathogen invasion, commensal bacterial colonization, antimicrobe peptide secretion, and mucosal defense…. The involvement of Vitamin D/VDR in anti-inflammation and anti-infection represents a newly identified and highly significant activity for VDR.

Jun Sun 7

When activated by 1,25-D, the Vitamin D Receptor (also called the calcitriol receptor) transcribes thousands of genes.8 It is commonly known that the VDR functions in regulating calcium metabolism.9 It is becoming increasingly clear, however, that the clinically accepted role of the Vitamin D metabolites, that of regulating calcium homeostasis, is just a small subset of the functions actually performed by these hormones. 

Transcription of antimicrobial peptides

One of the VDR’s key functions is the transcription of antimicrobial peptides.10 11 See below.  

Other antimicrobial activity of the VDR

Additionally, when the VDR is activated, TLR2 is expressed.12 TLR2 is a receptor, which is expressed on the surface of certain cells and recognizes native or foreign substances, and passes on appropriate signals to the cell and/or the nervous system.

When activated TLR2 allows the immune system to recognize gram-positive bacteria, including Staphylococcus aureus13 14Chlamydia pneumoniae15 and Mycoplasma pneumoniae.16 TLR2 also protects from intracellular infections such as Mycobacteria tuberculosis.17  

Antimicrobial peptides

The antimicrobial peptides (AMPs), of which there are hundreds, are families of proteins, which have been called “the body’s natural antibiotics,” crucial for both prevention and clearance of infection. AMPs are broad-spectrum, responding to pathogens in a non-specific manner.18

For example, consider cathelicidin, a protein transcribed the VDR, which not unlike a Swiss Army knife, has many different functions. Because it can be differentially spliced, the cathelicidin protein itself can respond to a range of very different microbial challenges. In humans, the cathelicidin antimicrobial peptide gene encodes an inactive precursor protein (hCAP18) that is processed to release a 37amino-acid peptide (LL-37) from the C-terminus. LL-37 is susceptible to proteolitic processing by a variety of enzymes, generating many different cathelicidin-derived peptides, each of which has specific targets. For example, LL-37 is generated in response toStaphylococcus aureus, yet LL-37 represents 20% of the cathelicidin-derived peptides, with the smaller peptides being much more abundant and able to target even more diverse microbial forms.19

AMPs have been documented to kill bacteria and disrupt their function through the following modes of action:

  • interfering with metabolism
  • targeting cytoplasmic components
  • disrupting membranes
  • act as chemokines and/or induce chemokine production, which directs traffic of bacteria

Also, AMPs aid in recovery from infection by:

  • promoting wound healing
  • inhibiting inflammation

In many cases, the exact mechanism by which antimicrobial peptides kill bacteria is unknown. In contrast to many conventional antibiotics including those used by the Marshall Protocol, AMPs appear to be bacteriocidal (a killer of bacteria) instead of bacteriostatic (an inhibitor of bacterial growth).

Two of the more significant families of AMPs are cathelicidin and the beta-defensins. Of these two families, cathelicidin is the most common.

The full extent by which microbes interfere with AMP expression is the subject of a rapidly growing body of research.20 21 22

Antimicrobial peptides target fungi and viruses

The antimicrobial peptides play a role in mitigating the virulence of the virome and other non-bacterial infectious agents. In addition to its antibacterial activity, alpha-defensin human neutrophil peptide-1 inhibits HIV and influenza virus entry into target cells.23 It diminishes HIV replication and can inactivate cytomegalovirus, herpes simplex virus, vesicular stomatitis virus and adenovirus.24 In addition to killing both gram positive and gram-negative bacteria, human beta-defensins HBD-1, HDB-2, and HBD-3 have also been shown to kill the opportunistic yeast species Candida albicans.25 Cathelicidin also possesses antiviral and antifungal activity.26 27

In other words, there is a reason why this group of proteins are named antimicrobial peptides rather than antibacterial peptides.

Unexpected antimicrobial peptides

There are now several examples of substances believed to cause disease, which have since been proven to be part of host defense.

  • amyloid beta (amyloid-β) – In a seminal 2010 study, a team of Harvard researchers showed that amyloid beta – the hallmark of Alzheimer’s disease – can act as an antimicrobial peptide, having antimicrobial activity against eight common microorganisms, including Streptococcus, Staphylococcus aureus, and Listeria.28 This led study author Rudolph E. Tanzi, PhD to conclude that amyloid beta is “the brain’s protector.” However, a 2010 study suggests that toxic levels of amyloid beta “dramatically suppresses VDR expression.” This suggests that overexpression of amyloid beta serves the interests of at least some microbes.29Read more.
  • certain human prion proteins   

Evolutionarily conserved

The TLR2/1 and cathelicidin-vitamin D pathway has long played a “powerful force” in protecting the body against infection. This is evidenced by the fact that the Alu short interspersed element (SINE), which transcribes the vitamin D receptor binding element (VDRE), has been evolutionarily conserved for 55-60 million years, but not prior.30 The differences in this pathway between humans/primates and other mammals call into question animal models that try to emulate the vitamin D system and indeed the immune system.

Inflammation

Another component of the innate immune response is inflammation, the universal initial response of the organism to any injurious agent.31 Inflammation is a systemic physiological process fundamental for survival.32 The identification of bacteria and other pathogens triggers the release of inflammatory cytokines. These cytokines include interferon-gamma, tumor necrosis factor-alpha (TNF-alpha), and Nuclear Factor-kappa B (NF-kappaB). Cytokines are regulatory proteins, such as the interleukins and lymphokines, that are released by cells of the immune system and act as intercellular mediators in the generation of an immune response. The result is what medicine calls inflammation, which generally leads to an increase in symptoms.

Th1/Th17 inflammation

One key type of inflammation is the Th1/Th17 (T-helper) inflammatory response. In the interests of concision, the Th1/Th17, on this site and others, the Th1/Th17 response is referred to as the Th1 response. This reaction occurs in response to intracellular pathogens, which according to the Marshall Pathogenesis, play a driving force in chronic disease.

All Th1 diseases are marked by an inflammatory response

Before the Human Microbiome Project, scientists couldn’t consistently link bacteria to inflammatory diseases. But with the advent of DNA sequencing technology, scientists have detected many of the bacteria capable of generating an inflammatory response. All diseases of unknown etiology are inflammatory diseases.

An inflammatory immune response—one of the body’s primary means to protect against infection—defines multiple established infectious causes of chronic diseases, including some cancers. Inflammation also drives many chronic conditions that are still classified as (noninfectious) autoimmune or immune-mediated (e.g., systemic lupus erythematosus, rheumatoid arthritis, Crohn’s disease). Both [the innate and adaptive immune systems] play critical roles in the pathogenesis of these inflammatory syndromes. Therefore, inflammation is a clear potential link between infectious agents and chronic diseases.

Siobhán M. O’Connor et al. 33

Th2 inflammation

According to the Marshall Pathogenesis, generally speaking, any activity of the Th2 cytokines in chronic disease is a result of the primary Th1-inducing pathogens.

Many palliative therapies interfere with inflammation

While inflammation is associated with disease, inflammation often serves an invaluable role as the immune system fights off chronic pathogens. Numerous medications artificially suppress inflammation including anti-TNF drugs, interferon, corticosteroids, antifungals, and anti-pyreutics. While interfering with the inflammatory response typically reduces immunopathology and makes a patient feel less symptomatic in the near term, doing so allows the bacteria which cause chronic disease to proliferate.

The release of cytokines appears to be essential for recovery after an infection. One study found that the cytokine TNF-alpha – which is blocked by anti-TNF drugs – is necessary for the proper expression of acquired specific resistance following infection withMycobacterium tuberculosis.34 35 36 Another effect of the use of TNF blockers is to break or reduce the formation of granuloma, one of the body’s mechanisms to control bacterial pathogens.37

Commensal microbes

The host innate immune defense system is highly active in healthy tissue.38 Commensal bacteria can activate innate immune responses.39 40

Keywords:
References
2 Ramagopalan SV, Heger A, Berlanga AJ, Maugeri NJ, Lincoln MR, Burrell A, Handunnetthi L, Handel AE, Disanto G, Orton SM, Watson CT, Morahan JM, Giovannoni G, Ponting CP, Ebers GC, Knight JC A ChIP-seq defined genome-wide map of vitamin D receptor binding: associations with disease and evolution. Genome Res. 2010;20:1352-60.
3 , 8 Wang TT, Tavera-Mendoza LE, Laperriere D, Libby E, MacLeod NB, Nagai Y, Bourdeau V, Konstorum A, Lallemant B, Zhang R, Mader S, White JH Large-scale in silico and microarray-based identification of direct 1,25-dihydroxyvitamin D3 target genes. Mol Endocrinol. 2005;19:2685-95.
4 , 18 Zasloff M Antimicrobial peptides of multicellular organisms. Nature. 2002;415:389-95.
6 Overington JP, Al-Lazikani B, Hopkins AL How many drug targets are there? Nat Rev Drug Discov. 2006;5:993-6.
7 Sun J Vitamin D and mucosal immune function. Curr Opin Gastroenterol. 2010;:.
9 Li YC, Bolt MJ, Cao LP, Sitrin MD Effects of vitamin D receptor inactivation on the expression of calbindins and calcium metabolism. Am J Physiol Endocrinol Metab. 2001;281:E558-64.
10 Wang TT, Nestel FP, Bourdeau V, Nagai Y, Wang Q, Liao J, Tavera-Mendoza L, Lin R, Hanrahan JW, Mader S, White JH Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. J Immunol. 2004;173:2909-12.
12 Schauber J, Dorschner RA, Coda AB, Büchau AS, Liu PT, Kiken D, Helfrich YR, Kang S, Elalieh HZ, Steinmeyer A, Zügel U, Bikle DD, Modlin RL, Gallo RL Injury enhances TLR2 function and antimicrobial peptide expression through a vitamin D-dependent mechanism. J Clin Invest. 2007;117:803-11.
14 González-Zorn B, Senna JP, Fiette L, Shorte S, Testard A, Chignard M, Courvalin P, Grillot-Courvalin C Bacterial and host factors implicated in nasal carriage of methicillin-resistant Staphylococcus aureus in mice. Infect Immun. 2005;73:1847-51.
15 Cao F, Castrillo A, Tontonoz P, Re F, Byrne GI Chlamydia pneumoniae–induced macrophage foam cell formation is mediated by Toll-like receptor 2. Infect Immun. 2007;75:753-9.
16 Chu HW, Jeyaseelan S, Rino JG, Voelker DR, Wexler RB, Campbell K, Harbeck RJ, Martin RJ TLR2 signaling is critical for Mycoplasma pneumoniae-induced airway mucin expression. J Immunol. 2005;174:5713-9.
17 Carlos D, Frantz FG, Souza-Júnior DA, Jamur MC, Oliver C, Ramos SG, Quesniaux VF, Ryffel B, Silva CL, Bozza MT, Faccioli LHTLR2-dependent mast cell activation contributes to the control of Mycobacterium tuberculosis infection. Microbes Infect.2009;11:770-8.
20 Chakraborty K, Ghosh S, Koley H, Mukhopadhyay AK, Ramamurthy T, Saha DR, Mukhopadhyay D, Roychowdhury S, Hamabata T, Takeda Y, Das S Bacterial exotoxins downregulate cathelicidin (hCAP-18/LL-37) and human beta-defensin 1 (HBD-1) expression in the intestinal epithelial cells. Cell Microbiol. 2008;10:2520-37.

 

Role of Dihydroxyvitamin D3 and Its Nuclear Receptor in Novel Directed Therapies for Cancer

S. Ondková, D. Macejová and J. Brtko
Gen. Physiol. Biophys. (2006), 25, 339—353   http://www.gpb.sav.sk/2006_04_339.pdf

Dihydroxyvitamin D3 is known to affect broad spectrum of various biochemical and molecular biological reactions in organisms. Research on the role and function of nuclear vitamin D receptors (VDR) playing a role as dihydroxyvitamin D3 inducible transcription factor belongs to dynamically developing branches of molecular endocrinology. In higher organisms, full functionality of VDR in the form of heterodimer with nuclear 9-cis retinoic acid receptor is essential for biological effects of dihydroxyvitamin D3. This article summarizes selected effects of biologically active vitamin D3 acting through their cognate nuclear receptors, and also its potential use in therapy and prevention of various types of cancer.

……

Vitamin D family consists of 9,10-secosteroids which differ in their side-chain structures. They are classified into five forms: D2, ergocalciferol; D3, cholecalciferol; D4, 22,23-dihydroergocalciferol; D5, sitosterol (24-ethylcholecalciferol) and D6, stigmasterol (Napoli et al. 1979). The main forms are vitamin D2 (ergocalciferol: plant origin) and vitamin D3 (cholecalciferol: animal origin). Both 25-hydroxyvitamin D2 and 1α,25-dihydroxyvitamin D2 have been evaluated for their biological functions. Vitamin D itself is a prohormone that is metabolically converted to the biologically active metabolite, 1,25-dihydroxyvitamin D3 in kidney. This vitamin D3, currently considered a steroid hormone, activates its cognate nuclear receptor (vitamin D receptor or VDR) which alter transcription rates of the target genes responsible for its biological responses. In general, vitamin D is essential for mineral homeostasis, for absorption and utilization of both calcium and phosphate and it aids in the mobilization of bone calcium and maintenance of serum calcium concentrations. Through these function, it plays an important role in ensuring proper functioning of muscles, nerves, blood clotting, cell growth and energy utilization. It has been proposed that vitamin D is also important for insulin and prolactin secretion, immune and stress responses, melanin synthesis and for differentiation of skin and blood cells (Lips 2006). Vitamin D metabolites also play a role in the prevention of auto-immune diseases and cancer (Pinette et al. 2003; Dusso et al. 2005). The steroid hormone 1α,25-dihydroxyvitamin D3 (calcitriol) exerts biological responses by interaction with both the well-characterized nuclear receptor (VDRnuc) responsible for activation gene transcription and not fully characterized membrane-associated protein/receptor (VDRmem) involved in generating a variety of rapid, non-genotropic responses (Evans 1988; Norman et al. 2002).

Vitamin D metabolism

Vitamin D, the “sunshine” vitamin, is synthesized under the influence of ultraviolet light in the skin. Many mammals have provitamin D (7-dehydrocholesterol) which is converted to provitamin D3 in their skin. When human skin is exposed to sunlight, the UV-B photons (wavelengths 290–315 nm) interact with 7-dehydrocholesterol causing photolysis and cleavage of the B-ring of the steroid structure, which upon thermoisomerization yields a secosteroid. Thus, provitamin D3 which is inherently unstable rapidly converts by a temperature-dependent process to vitamin D3 (MacLaughlin et al. 1982; Holick 1994). Vitamin D3 enters the blood circulation and binds to vitamin D binding protein (DBP) (Haddad et al. 1993) which carries vitamin D3 to liver and kidney for bioactivation (Wikvall 2001). In the first activation step, vitamin D3 is hydroxylated by the enzyme 25-hydroxylase to 25- hydroxyvitamin D3 mainly in the liver. This metabolite is present in the circulation at the concentration of more than 0.05 µmol/l (20 ng/ml). In the second step, the biologically active hormone 1α,25-dihydroxyvitamin D3 is generated by hydroxylation of 25-hydroxyvitamin D3 at 1α-position in kidney. The enzyme 1α-hydroxylase has been shown to be also present in keratinocytes and prostate epithelial cells, suggesting that those organs may also be able to generate 1α,25-dihydroxyvitamin D3 from 25-dihydroxyvitamin D3 (Schwartz et al. 1998). The activity of 1α-hydroxylase in the kidney serves as the major control point in production of the active hormone. The active metabolite 1α,25-dihydroxyvitamin D3 is present in human plasma at the concentration ranging from 0.05 to 0.15 nmol/l (20–60 pg/ml) (Hartwell et al. 1987; Gross et al. 1996). In general, 90 to 100% of the most human being vitamin D requirement comes from exposure to sunlight (Holick 2003) and the rest of the vitamin D3 content is obtained from diet (Malloy and Feldman 1999). The catabolism of vitamin D occurs by further hydroxylation of 25-dihydroxyvitamin D3 by 24-hydroxylase to yield 24,25-dihydroxyvitamin D3. The 24-hydroxylase is ubiquitous enzyme and is expressed in all the cells expressing VDR. This enzyme is regulated by parathyroid hormone and 1α,25-dihydroxyvitamin D3. The major significance of 24-hydroxylation is inactivation of vitamin D (Nishimura et al. 1994; Brenza and DeLuca 2000). The combinations of 1,25-dihydroxyvitamin D3 with inhibitors of 24-hydroxylase such as ketoconazole or liarozole may enhance its antitumour effects in prostate cancer therapy.

Vitamin D3 receptor

More than 2000 synthetic analogues of the biological active form of vitamin D, 1α,25-dihydroxyvitamin D3, are presently known. Basically, all of them interfere with the molecular switch of nuclear 1α,25-dihydroxyvitamin D3 signalling, which is the complex of the VDR, the retinoid X receptor (RXR), and a 1α,25-dihydroxyvitamin D3 response element (VDRE) (Carlberg 2003).

VDR is the only nuclear protein that binds the biologically most active vitamin D metabolite, 1α,25-dihydroxyvitamin D3, with high affinity (Kd = 0.1 nmol/l). This classifies the VDR into the classical endocrine receptor subgroup of the nuclear receptor superfamily, which also contains the nuclear receptors for hormones as retinoic acid, thyroid hormone, estradiol, progesterone, testosterone, cortisol, and aldosterol (Carlberg 1995). Similarly, like other biologically active ligand for nuclear hormone receptors, 1,25-dihydroxyvitamin D3 can modulate expression of selected ion transport protein genes (Van Baal et al. 1996; Hudecova et al. 2004).

The VDR was first isolated after trancfection of COS-1 cells with cloned sequences of complementary DNA that was isolated from human intestine (Baker et al. 1988). VDR has been found in more than 30 tissues including intestine, colon, breast, lung, ovary, bone, kidney, parathyroid gland, pancreatic β-cells, monocytes, keratinocytes, and many cancer cells, suggesting that the vitamin D endocrine system may also be involved in regulating the immune systems, cellular growth, differentiation and apoptosis (Jones et al. 1998). The active form of vitamin D binds to intracellular receptors that then function as transcription factors to modulate gene expression. Like the receptors for other steroid hormones and thyroid hormones, the VDR has specific hormone-binding and DNA-binding domains. It contains two zinc finger structures forming a characteristic DNA-binding domain (DBD) of 66 amino acids and a carboxy-terminal ligand-binding domain (LBD) of approximately 300 amino acids, which is formed by 12 α-helices. Ligand binding causes a conformational change within the LBD, in which helix 12, the most carboxy-terminal α-helix, closes the ligand-binding pocket via a “mouse-trap like” intramolecular folding (Moras and Gronemeyer 1998). Moreover, the LBD is involved in a variety of interactions with nuclear proteins, such as other nuclear receptors, corepressor and coactivator proteins. These ligand-triggered protein-protein interactions are the central molecular event of nuclear 1α,25-dihydroxyvitamin D3 signalling.

….

Role of vitamin D3 in cancer

Some of biologically active ligands for nuclear receptors exert tumour-suppressive activity, and they have therapeutical exploitation due to their antiproliferative and apoptosis-inducing effects (Brtko and Thalhamer 2003).

…..

During the last decade, evidence for vitamin D3 effects has been accumulating not only for prostate cancer (Feldman et al. 1995; Ma et al. 2004) but also for colon cancer (Cross et al. 1997; Bischof et al. 1998). 1α-hydroxylase was found to be Vitamin D and Cancer Treatment 347 expressed and active in colorectal cancer (Bareis et al. 2001; Cross et al. 2001; Tangpricha et al. 2001; Ogunkolade et al. 2002) and ovarian cancer (Miettinen et al. 2004). In both colon and also lung tumours, CYP24A1 mRNA was significantly up-regulated, while VDR mRNA was generally down-regulated when compared to respective normal tissues. When the level of VDR in 12 malignant colonic tumours was compared with that of adjacent normal tissue, in 9 cases out of 12, expression of VDR in tumours was decreased. However, in that study, the expression of CYP24A1 was not assessed. It has also been shown that, at least in human colon cancer cell lines, the level of VDR correlates with the degree of cell differentiation (Shabahang et al. 1993; Anderson et al. 2006).

Recently, it has been suggested that actually 20–30% of colorectal cancer incidence might be due to insufficient exposure to sunlight. This fact was strengthen by correlation between reduced colorectal cancer incidence and sunlight exposure, low skin pigmentation, nutritional vitamin D intake and high serum levels of 25- hydroxyvitamin D3 (Grant and Garland 2003). In the colon at least, CYP27B1 and VDR expression was described to be actually elevated during early tumour progression and that described dual positivity was found in many, but not all the tumour cells. In human colon tumours, CYP24 mRNA is quite highly expressed and the studies also demonstrated that with the exception of differentiated Caco-2 cells, CYP24 activity is constitutively present or can be induced by 1α,25- dihydroxyvitamin D3. During tumour progression in the colon, not only VDR but CYP27B1 and CYP24 expression were found to be increased in tumour tissues (Bareis et al. 2001; Bises et al. 2004).

Androgens, retinoids, glucocorticoids, estrogens and agonists of peroxisome proliferator-activated receptor directly or indirectly have reasonable impact on vitamin D signalling pathways, and vice versa. It was proposed that sex hormones might reduce colorectal cancer risk (McMichael and Potter 1980). The studies suggested that current and long-term use of estrogens is associated with a substantial decrease in risk of fatal colon cancer. The mechanism, however, by which estrogens could inhibit colonic tumour growth, remains an enigma. There are at least two distinct estrogen receptors in the human body: ERα and ERβ. In the normal human colon, ERβ is widely regarded to be the predominant subtype (CampbellThompson et al. 2001). In a recently terminated pilot study together with Strang Cancer Prevention Centre at Rockefeller University (NY, USA), tissues from postmenopausal women receiving 17β-estradiol for expression of CYP27B1 by real time RT-PCR were examined. CYP27B1 was found to be elevated significantly in all subjects after receiving 17β-estradiol for 4 weeks.

Amplification of chromosomal region 20q12q13 containing the CYP24A1 gene has been reported in ovarian cancer, as well (Tanner et al. 2000). Although inhibition of ovarian cancer cell growth by 1α,25-dihydroxyvitamin D3 has been reported (Saunders et al. 1992, 1995), a clinical trial testing the efficacy of 1α,25- dihydroxyvitamin D3 combined with isotretinoin in treating 22 epithelial ovarian cancer patients for 74 weeks has not produced positive results (Rustin et al. 1996).

Read Full Post »

Lonely Receptors: RXR – Jensen, Chambon, and Evans

Larry H. Bernstein, MD, FCAP, Curator

Leaders in Pharmaceutical Intelligence

Series E. 2; 7.2

 

Nuclear receptors provoke RNA production in response to steroid hormones

Albert Lasker Basic Medical Research Award

Pierre Chambon, Ronald Evans and Elwood Jensen

For the discovery of the superfamily of nuclear hormone receptors and elucidation of a unifying mechanism that regulates embryonic development and diverse metabolic pathways.

Hormones control a vast array of biological processes, including embryonic development, growth rate, and body weight. Scientists had known since the early 1900s that tiny hormone doses dramatically alter physiology, but they had no idea that these signaling molecules did so by prodding genes. The 1950s, when Jensen began his work, was the great era of enzymology. Conventional wisdom held that estradiol—the female sex hormone that instigates growth of immature reproductive tissue such as the uterus—entered the cell and underwent a series of chemical reactions that produced a particular compound as a byproduct. This compound—NADPH—is essential for many enzymes’ operations but its small quantities normally limit their productivity. A spike in NADPH concentrations would stimulate growth or other activities by unleashing the enzymes, the reasoning went.

In 1956, Jensen (at the University of Chicago) decided to scrutinize what happened to estradiol within its target tissues, but he had a problem: The hormone is physiologically active in minute quantities, so he needed an extremely sensitive way to track it. He devised an apparatus that tagged it with tritium—a radioactive form of hydrogen—at an efficiency level that had not previously been achieved. This innovation allowed him to detect a trillionth of a gram of estradiol.

When he injected this radioactive substance into immature rats, he noticed that most tissues—skeletal muscle, kidneys and liver, for example—started expelling it within 15 minutes. In contrast, tissues known to respond to the hormone—those of the reproductive tract—held onto it tightly. Furthermore, the hormone showed up in the nuclei of cells, where genes reside. Something there was apparently grabbing the estradiol.

Jensen subsequently showed that his radioactive hormone remained chemically unchanged once inside the cell. Estrogen did not act by being metabolized and producing NADPH, but presumably by performing some job in the nucleus. Subsequent work by Jensen and Jack Gorski established that estradiol converts a protein in the cytoplasm, its receptor, into a form that can migrate to the nucleus, embrace DNA, and turn on specific genes.

From 1962 to 1980, molecular endocrinologists built on Jensen’s work to discover the receptors for the other major steroid hormones—testosterone, progesterone, glucocorticoids, aldosterone, and the steroid-like vitamin D. In addition to Jensen and Gorski, many scientists—notably Bert O’Malley, Jan-Ake Gustafsson, Keith Yamamoto, and the late Gordon Tompkins—made crucial observations during the early days of steroid receptor research.

Clinical Applications of Estrogen-Receptor Detection

Clinicians knew that removing the ovaries or adrenal glands of women with breast cancer would stop tumor growth in one out of three patients, but the molecular basis for this phenomenon was mysterious. Jensen showed that breast cancers with low estrogen-receptor content do not respond to surgical treatment. Receptor status could therefore indicate who would benefit from the procedure and who should skip an unnecessary operation. In the mid-1970s, Jensen and his colleague Craig Jordan found that women with cancers that contain large amounts of estrogen receptor are also likely to benefit from tamoxifen, an anti-estrogen compound that mimics the effect of removing the ovaries or adrenal glands. The other patients—those with small numbers of receptors—could immediately move on to chemotherapy that might combat their disease rather than waiting months to find out that the tumors were growing despite tamoxifen treatment. By 1980, Jensen’s test had become a standard part of care for breast cancer patients.

In the meantime, Jensen set about generating antibodies that bound the receptor—a tool that provided a more reliable way to measure receptor quantities in excised breast tumor specimens. His work has transformed the treatment of breast cancer patients and saves or prolongs more than 100,000 lives annually.

Long-Lost Relatives

By the early 1980s, interest in molecular endocrinology had shifted toward the rapidly developing area of gene control. Chambon and Evans had long wondered how genes turn on and off, and recognized nuclear hormone signaling as the best system for studying regulated gene transcription. They wanted to know exactly how nuclear receptors provoke RNA production in response to steroid hormones. To manipulate and analyze the receptors, they would need to isolate the genes for them.

By late 1985 and early 1986, Evans (at the Salk Institute in La Jolla) and Chambon (at the Institute of Genetics and Molecular and Cellular Biology in Strasbourg, France) had pieced together the glucocorticoid and estrogen receptor genes, respectively. They noticed that the sequences resembled that of v-erbA, a miscreant viral protein that fosters uncontrolled cell growth. This observation raised the possibility that v-erbA and its well-behaved cellular counterpart, c-erbA, would also bind DNA and control gene activity in response to some chemical activator, or ligand. In 1986, Evans and Björn Vennström simultaneously reported that c-erbA was a thyroid hormone receptor that was related to the steroid hormone receptors, thus uniting the fields of thyroid and steroid biology.

Chambon and Evans set to work deconstructing the glucocorticoid and estrogen receptors. By creating mutations at different spots and probing which activities the resulting proteins lost, they dissected the receptor into three domains: one bound hormone, one bound DNA, and one activated target genes. The structure of each domain strongly resembled the analogous one in the other receptor.

Chambon and Evans wanted to match other members of the growing receptor gene family with their chemical triggers. Because the DNA- and ligand-binding regions functioned independently, it was possible to hook the DNA-binding domain of, say, the glucocorticoid receptor to the ligand-binding domain of another receptor whose ligand was unknown. The ligand for that receptor would then activate a glucocorticoid-responsive test gene.

Evans would use this method to identify ligands for several novel members of the nuclear receptor family, and both he and Chambon exploited it to discover a physiologically crucial receptor. In the late 1970s, scientists had suggested that the physiologically active derivative of vitamin A, retinoic acid, could exert its effects by binding to a nuclear receptor. This nutrient is essential from fertilization through adulthood, and researchers were eager to understand its activities on a molecular level. During embryonic development, deficiency of retinoic acid impairs formation of most organs, and the compound can hinder cancer cell proliferation. So Chambon set out to find a receptor that responded to retinoic acid. He isolated a member of the nuclear receptor gene family whose production increased in breast cancer cells that slowed their growth upon exposure to the chemical. Simultaneously, Evans identified the same protein. He tested whether more than a dozen compounds activated an unknown receptor and one passed: retinoic acid.

Remarkably, in 1986, the two scientists had independently—and unbeknownst to each other—identified the same retinoic acid receptor, a molecule of tremendous significance. The discovery of this molecule provided an entry point for detailing vitamin A biology.

Rx for Lonely Receptors: RXR

The list of presumptive nuclear receptors was growing quickly as scientists realized that the common DNA sequences provided a handle with which to grab these molecules from the genome. Because their chemical activators weren’t known, they were called “orphan” receptors, and researchers were keen on “adopting” them to ligands. Some of these ligands, they reasoned, would represent previously unknown classes of gene activators. The test system that Chambon and Evans used to match up retinoic acid with its receptor, in which they stitched an unknown ligand-binding domain to a DNA-binding domain for a receptor with known target sequences, could be harnessed to accomplish this task.

Evans had identified some potential nuclear receptors from fruit flies. He decided to pursue a human orphan receptor that closely resembled one of these receptor genes, reasoning that a protein that functioned in both flies and mammals was likely to perform an important job.

This receptor responded to retinoic acid in intact cells but did not bind it in the test tube, so Evans called it the Retinoid X Receptor (RXR), thinking that its ligand was some retinoic acid derivative. In cells, enzymes convert retinoic acid to metabolites and it seemed possible that one of these compounds was RXR’s ligand. In 1992, Evans’s group and one at Hoffmann-La Roche discovered that 9-cis-retinoic acid, a stereoisomer of retinoic acid, could activate RXR, identifying the first new receptor ligand in 25 years. This finding launched the orphan receptor field because it provided strong evidence that the strategy could unearth previously unknown ligands.

In the meantime, Chambon had found that the purified retinoic acid receptor, in contrast to the estrogen receptor, did not bind efficiently to its target DNA. Other nuclear receptors, too, needed help grasping genes. In the test tube, the retinoic acid, thyroid hormone, and vitamin D3 receptors could attach well to their target DNA only when supplemented with cellular material, which presumably contained some crucial substance. Chambon and Michael Rosenfeld independently purified a single protein that performed this feat, and it turned out to be none other than RXR. This ability of RXR to pair with other receptors—forming so-called heterodimers—would turn out to be key for switching on many orphan receptors. These heterodimeric couplings yield large numbers of distinct gene-controlling entities.

Chambon revealed the power of mixing and matching in these molecular duos through his thorough and extensive genetic manipulations in mice. He has shown that vitamin A exerts its wide-ranging effects on organ development in the embryo through the action of eight different forms of the retinoic acid receptor and six different forms of RXR, interacting with each other in a multitude of combinations.

Clinical Applications of the Superfamily Work

The concept of RXR as a promiscuous heterodimeric partner for certain nuclear receptors led to the unexpected identification of a number of clinically relevant receptors. These proteins include the peroxisome proliferator-activated receptor (PPAR), which stimulates fat-cell maturation and sits at the center of Type 2 diabetes and a number of lipid-related disorders; the liver X receptors (LXRs) and bile acid receptor (FXR), which help manage cholesterol homeostasis; and the steroid and xenobiotic receptor (PXR), which turns on enzymes that dispose of chemicals that need to be detoxified, such as drugs.

Because the nuclear receptors wield such physiological power, they have provided excellent targets for disease treatment. The anti-diabetes compounds glitazones, for example, work by stimulating PPAR, and the clinically used lipid-lowering medications called fibrates work by binding a closely related receptor, PPAR. Retinoic acid therapy has dramatically altered the prognosis of people with acute promyelocytic leukemia by triggering specialization of the immature white blood cells that accumulate in these individuals. The three-dimensional structure of nuclear receptors with and without their ligands, which Chambon and his colleagues first solved, promises to accelerate drug discovery in the whole field.

Nuclear hormone receptors have touched on human health in other ways as well. Genetic perturbations in the genes for these proteins cause a variety of illnesses. For example, certain forms of rickets arise from mutations in the vitamin D receptor and several disorders of male sexual differentiation stem from defects in the androgen receptor.

The discoveries of Jensen, Chambon, and Evans revealed an unimagined superfamily of proteins. At the start of this work almost 50 years ago, no one would have anticipated that steroids, thyroid hormone, retinoids, vitamin D, fatty acids, bile acids, and many lipid-based drugs transmit their signal through similar pathways. Four dozen human nuclear receptors are now known, and scientists are working out the roles of these proteins in normal and aberrant physiology. These discoveries have revolutionized the fields of endocrinology and metabolism, and pointed toward new tactics for drug discovery.

by Evelyn Strauss, Ph.D.

 

The 2004 Lasker Award for Basic Medical Research will be presented to Elwood Jensen, Ph.D., the Charles B. Huggins Distinguished Service Professor Emeritus in the Ben May Institute for Cancer Research at the University of Chicago, one of three scientists whose discoveries “revolutionized the fields of endocrinology and metabolism,” according to the award citation. Jensen’s work had a rapid, direct and lasting impact on treatment and prevention of breast cancer.

The Lasker Awards are the nation’s most distinguished honor for outstanding contributions to basic and clinical medical research. Often called “America’s Nobels,” the Lasker Award has been awarded to 68 scientists who subsequently went on to receive the Nobel Prize, including 15 in the last 10 years.

Jensen will share the basic medical research award with two colleagues, Pierre Chambon, of the Institute of Genetics and Molecular and Cellular Biology (Strasbourg, France), and Ronald M. Evans of the Salk Institute for Biological Studies (La Jolla, California) and the Howard Hughes Medical Institute.

They were selected for their discovery of the “superfamily of nuclear hormone receptors and the elucidation of a unifying mechanism that regulates embryonic development and diverse metabolic pathways.” The implications of this research for understanding human disease and accelerating drug discovery “have been profound and hold much promise for the future,” notes the announcement from the Lasker Foundation.

Jensen is being honored for his pioneering research on how steroid hormones, such as estrogen, exert their influence. His discoveries explained how these hormones work, which has led to the development of drugs that can enhance or inhibit the process.

Hormones control a vast array of biological processes, including embryonic development, growth rate and body weight. Before Jensen, however, the way which hormones cause these effects was “a complete mystery,” recalled Gene DeSombre, Ph.D., professor emeritus at the University of Chicago, who worked with Jensen in the Ben May Institute as a post-doctoral fellow and then as a colleague.

In the 1950s, biochemists thought a hormone entered a cell, where a series of oxidation and reductions reactions with the estrogen provided needed energy for the growth stimulation and other specific actions shown by estrogens.

From the late 1950s to the 1970s Jensen entirely overturned that notion. Working with estrogen, he proved that hormones do not undergo chemical change. Instead, they bind to a receptor protein within the cell. This hormone-receptor complex then travels to the cell nucleus, where it regulates gene expression.

At the time, this idea was heresy. “That really got him into some hot water,” recalled DeSombre. “Jensen struggled quite a lot,” echoes Shutsung Liao, Ph.D., another Ben May colleague, who subsequently found a similar system for testosterone action. But for Jensen, just getting into hot water was a struggle. When he first presented preliminary data at a 1958 meeting in Vienna, only five people attended, three of whom were the other speakers. More than 1,000 attended a simultaneous symposium on the metabolic processing of estrogen.

In the next 20 years, Jensen convinced his colleagues by publishing a series of major and highly original discoveries in four related areas of hormone research:

  • Jensen discovered the estrogen receptor, the first receptor found for any hormone. In 1958, using a radioactive marker, he showed that only the tissues that respond to estrogen, such as those of the female reproductive tract, were able to concentrate injected estrogen from the blood. This specific uptake suggested that these cells must contain binding proteins, which he called “estrogen receptors.”
  • In 1967, Jensen and Jack Gorski of the University of Wisconsin showed that these putative receptors were macromolecules that could be extracted from these tissues. With this method, Jensen showed that when estrogen bound to this receptor, the compound then migrated to the nucleus where it bound avidly and activated specific genes, stimulating new RNA synthesis.
  • By 1968, Jensen had devised a reliable test for the presence of estrogen receptors in breast cancer cells. It had been known for decades that about one-third of premenopausal women who had advanced breast cancer would respond to estrogen blockade brought about by removing their ovaries, the source of estrogen, but there was no way to predict which women would respond. In 1971, Jensen showed that women with receptor-rich breast cancers often have remissions following removal of the sources of estrogen, but cancers that contain few or no estrogen receptors do not respond to estrogen-blocking therapy.
  • By 1977, Jensen and Geoffrey Greene, Ph.D., also in the University of Chicago’s Ben May Institute, had developed monoclonal antibodies directed against estrogen receptors, which enabled then to quickly and accurately detect and count estrogen receptors in breast and other tumors. By 1980, this test had become a standard part of care for breast cancer patients

This work “transformed the treatment of breast cancer patients,” notes the Lasker Foundation, “and saves or prolongs more than a 100,000 lives annually.”

”Jensen’s revolutionary discovery of estrogen receptors is beyond doubt one of the major achievements in biochemical endocrinology of our time,” said DeSombre. “His work is hallmarked by great technical ingenuity and conceptual novelty. His promulgation of simple yet profound ideas concerning the role of receptors in estrogen action have been of the greatest importance for research on the basic and clinical physiology not only of estrogens but also of all other categories of steroid hormones.”

By the early 1970s, Jensen was searching for chemical, rather than surgical, ways to shield estrogen-dependent tumors from circulating hormones. He and colleague Craig Jordan (then at the Worcester Foundation for Experimental Biology in Massachusetts) subsequently found that women with cancers that contain large amounts of estrogen receptor are also likely to benefit from tamoxifen, a compound that blocks some of the effects of estrogen. Patients with few or no receptors could immediately move on to chemotherapy rather than waiting months to find out that the tumors were growing despite tamoxifen treatment.

Following Jensen’s lead, researchers soon found that the receptors for the other major steroid hormones, such as testosterone, progesterone, and cortisone, worked essentially the same way.

In 1986, Pierre Chambon and Ronald Evans separately but simultaneously discovered that the steroid hormone receptors were merely the tip of the iceberg of what would turn out to be a large family of structurally related nuclear receptors, now known to consist of 48 members. Evans and Chambon unearthed a number of these receptors, which revealed new regulatory systems that control the body’s response to essential nutrients (such as Vitamin A), fat-soluble signaling molecules (such as fatty acids and bile acids), and drugs (such as the glitazones used to treat Type 2 diabetes and retinoic acid for certain forms of acute leukemia).

These three individuals “created the field of nuclear hormone receptor research, which now occupies a large area of biological and medical investigation,” said Dr. Joseph L. Goldstein, chairman of the international jury of researchers that selects recipients of the Lasker Awards, and recipient of the Lasker Award for Basic Medical Research and the Nobel Prize in Medicine in 1985.

They revealed the “unexpected and unifying mechanism by which many signaling molecules regulate a plethora of key physiological pathways that operate from embryonic development through adulthood. They discovered a family of proteins that allows chemicals as diverse as steroid hormones, Vitamin A, and thyroid hormone to perform in the body.”

Jensen, known for concluding his lectures in verse, neatly summed up what his extraordinary series of discoveries might mean to a woman who has been diagnosed with breast cancer:

“A lady with growth neoplastic
Thought surgical ablation too drastic.
She preferred that her ill
Could be cured with a pill,
Which today is no longer fantastic.”

JBC THEMATIC MINIREVIEW SERIES 2011

Nuclear Receptors in Biology and Diseases

Thematic Minireview Series on Nuclear Receptors in Biology and Diseases

Sohaib Khan and Jerry B Lingrel

Although a connection between breast cancer and the ovary was made by Sir George Beatson in 1896 and estrogen was purified in 1920, it remained puzzling as to how the hormone exerted its biological effects. In the late 1950s, when Elwood Jensen delved into this problem by asking, essentially, “What does tissue do with this hormone?” little did he know that his quest would lead to the establishment of the nuclear receptor field. The late 1950s was the era of intermediary metabolism and enzymology, when steroid hormones were considered likely substrates in the formation of metabolites that functioned as cofactors in an essential metabolic pathway. The biological responses to estrogens and other steroids were thought to be mediated by enzymes. Against this background and prevailing dogma, Jensen and colleagues defined the biochemical mechanisms by which steroid hormones exert their effects. While working at the University of Chicago’s Ben May Institute for Cancer Research, they synthesized tritium-labeled estradiol and concurrently developed a new method to measure its uptake in biological material. These tools enabled them to determine the biochemical fate of physiological amounts of hormone. They discovered that the reproductive tissues of the immature rat contain characteristic hormone-binding components with which estradiol reacts to induce uterine growth without itself being chemically changed. From the close correlation between the inhibition of binding and inhibition of growth response, Jensen established that the binding substances were receptors. Thus, we saw the birth of the first member of the nuclear receptor family (known as the estrogen receptor). These findings stimulated the search for other physiological receptors, and the pioneering works by Pierre Chambon, Ronald Evans, Jan-Åke Gustafsson, Bert W. O’Malley, and Keith Yamamoto led to the discoveries of the glucocorticoid receptor (GR),2 progesterone receptor, retinoic acid receptor, and orphan receptors. In a rather short span of time, the nuclear receptor family has grown into a 49-member-strong “superfamily.” This is a family whose members, functioning as sequence-specific transcription factors, have defined the many intricacies of the mechanism of transcription. These ligand-dependent transcription factors generally possess similar “domain organizations,” of which the DNA-binding domain and the ligand-binding domain are critical in amplifying the hormonal signals via the receptor target genes. The nuclear receptor family is divided into four groups: (i) Group 1 is composed of steroid hormone receptors that control target gene transcription by binding as homodimers to response element (RE) palindromes; (ii) in Group 2, the nuclear receptors heterodimerize with retinoid X receptor and generally bind to direct repeat REs; (iii) Group 3 consists of those orphan receptors that function as homodimers and bind to direct repeat REs; and (iv) orphan receptors in Group 4 function as monomers and bind to single REs.

Since the early demonstration by Jack Gorski and Jensen that the estrogen receptor (ER) activates transcription, the nuclear receptor field has come a long way. In addition to the first cloning of the polymerase II transcription factors (GR and ER cDNAs), of note is the discovery of steroid receptor coactivators (SRCs), a truly major piece of the transcriptional jigsaw puzzle, described by the laboratories of O’Malley and Myles Brown. The induction of coactivators and corepressors in the transcriptional machinery has expanded tremendously our understanding of this complex process. We now know that ligand binding to the respective receptors triggers a fascinating chain of events, including the translocation of the receptors to the nucleus, ligand-induced changes in the receptor conformations, receptor dimerization, interaction with the target gene promoter elements, recruitment of coactivators (or corepressors), chromatin remodeling, and subsequent interaction with the polymerase II complex to initiate transcription.

By virtue of their abilities to regulate a myriad of human developmental and physiological functions (reproduction, development, metabolism), nuclear receptors have been implicated in a wide range of diseases, such as cancer, diabetes, obesity, etc. Not surprisingly, drug companies are spending billions of dollars to develop medicines for cancer and metabolic disorders that involve nuclear receptors. More than 50 years after the discovery of the ER, the scientific community owes Jensen and other founding members of the nuclear receptor family much gratitude, for they have taken us through a remarkable expedition filled with eureka moments to understand how hormones and other ligands function!

This thematic minireview series will cover a range of topics in the nuclear receptor field. The minireviews include the current studies of identifying subtypes of the GR. Different receptors arise from alternative mRNA splicing and from the use of different promoter start sites and post-translational modifications, such as phosphorylation. The series covers the physiological roles of the different GRs. The field of orphan nuclear receptors and the search for possible ligands also are reviewed. One minireview concentrates largely on the following nuclear receptors: peroxisome proliferator-activated receptor (PPAR) α, PPARγ, Rev-erbα, and retinoic acid receptor-related orphan receptor α. ERα was the first identified and has been studied the most, whereas ERβ has not been studied in the same detail. ERβ is very important, and one of the minireviews provides a summary of the new biological functions that are being ascribed to it. Also, the development of small molecule inhibitors for the ER will be considered. An important aspect of nuclear receptor function is how these receptors function in transcription. The role of transcriptional coactivators in nuclear receptor gene regulation will be reviewed as well as how signal amplification and interaction are involved in transcription regulation by steroids. The SRC/p160 family of coregulators includes SRC-1, SRC-2, and SRC-3, and the latter has been shown to act as an oncogene, particularly in breast cancer. Molecular analysis of its role in breast cancer progression and metastasis will be the focus of one of the minireviews. In addition, interactions of nuclear receptors with the genome will be reviewed, as will the role of the homeodomain protein HoxB13 in specifying the cellular response to androgens. Mining nuclear receptor cistromes and how nuclear receptors reset metabolism also will be considered. The association of nuclear receptors (e.g. PPARδ) with physiological functions, such as circadian rhythm and muscle functions, will also be addressed. Finally, the role of nuclear receptors in disease using the retinoid X receptor α/β knock-out and transgenic mouse model skin syndromes and asthma will be reviewed. These are diverse and important topics that are critical in understanding the regulation of nuclear receptors and the biological roles they play in normal function and disease.

The Nuclear Receptor Superfamily: A Rosetta Stone for Physiology

Ronald M. Evans
Howard Hughes Medical Institute, Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
Molecular Endocrinology 19(6):1429–143   http://dx.doi.org:/10.1210/me.2005-0046

In the December 1985 issue of Nature, we described the cloning of the first nuclear receptor cDNA encoding the human glucocorticoid receptor (GR) (1). In the 20 yr since that event, our field has witnessed a quantum leap by the subsequent discovery and functional elaboration of the nuclear receptor superfamily (2)—a family whose history is linked to the evolution of the entire animal kingdom and whose actions, by decoding the genome, span the vast diversity of biological functions from development to physiology, pathology, and treatment. A messenger is an envoy or courier charged with transmitting a communication or message. In one sense, the cloning of that first messenger (the GR) represented the completion of a prediction that began with Elwood Jensen’s characterization of the first steroid receptor protein (3) and continued with the pioneering work of others in the steroid receptor field (including Gorski, O’Malley, Gustafsson, and Yamamoto). Yet, like the discovery of the Rosetta stone in 1799, the revelation of the GR sequence heralded a completely unpredictable demarcation in the field, helping to solve mysteries unearthed nearly 100 yr ago as well as opening a portal to the future. The beginnings of the adventure lie in disciplines such as medicine and nutrition, which gave rise to the emergent field of endocrinology in the first half of the last century. The purification of chemical messengers ultimately known as hormones from organs and vitamins from foods spurred the study of these compounds and their physiologic effects on the body. At about the same time, the field of molecular biology was emerging from the disciplines of chemistry, physics, and their application to biological problems such as the structure of DNA and the molecular events surrounding its replication and transcription. It would not be until the late 1960s and 1970s that endocrinology and molecular biology would begin to intersect as the link between receptors and transcriptional control were being laid down. During this time, the work of Jensen (4) and Gorski (5) identified a high-affinity estrogen receptor (ER) that suggested an action in the nucleus. Gordon Tomkins and his associates (J. Baxter, G. Ringold, E. B. Thompson, H. Samuels, H. Bourne, and others) were one of the most creative forces studying glucocorticoid action (6). Concurrent work by O’Malley, Gustafsson, and Yamamoto provided further, important evidence supporting a link between steroid receptor action and transcription (see accompanying perspective articles in this issue of Molecular Endocrinology). But whereas the steroid hormone field continued to evolve in this direction, it is of interest to note that the mechanism of action of thyroid hormone and retinoids remained clouded and controversial until the eventual cloning of their receptors in the late 1980s. Likewise, no one had foreseen the possibility that other lipophilic molecules (like oxysterols, bile acids, and fatty acids) would also function through a similar mechanism, or that other steroid receptor-like proteins existed that would play an important role in transcriptional regulation of so many diverse pathways. Thus, the GR isolation in 1985 led to the concept of a hidden superfamily of receptors that in a very real way provided the needed molecular code to unravel the puzzle of physiologic homeostasis.

Unconventional Gene-Ology

The study of RNA tumor viruses was ascendant, and the concept that they evolved by pirating key signaling pathways greatly influenced my future studies. With this training, I went on to work with Jim Darnell at the Rockefeller University on adenovirus transcription, a model brought to the lab by Lennart Philipson. At the time, adenovirus was one of the best tools to study programmed gene expression in an animal cell. My sole focus was to localize the elusive major late promoter, which provided my first Nature paper (7). Ed Ziff, a newly hired assistant professor from Cambridge, brought innovative unpublished DNA and RNA sequencing techniques that, after much technical angst, allowed us to sequence the major late promoter and derive the structure of the first eukaryotic polymerase II promoter (8). This thrilling result convinced me that the problem of gene control could be solved at the molecular level. Our next goal, which I shared with Michael Harpold in the Darnell lab, was to translate the concepts developed around adenovirus into cellular systems. My model was to analyze the glucocorticoid and thyroid hormone regulation of the GH gene. Under the strict federal guidelines for newly approved recombinant DNA research, we cloned the GH cDNA in 1977 and the first genomic clones in 1978 (9) after I moved on to The Salk Institute. However, to fully address the hormone signaling problem, I realized that it would be necessary to clone the GR and thyroid hormone receptors (TRs), which began in earnest in 1981. Up until that time, the purification and cloning of any polymerase II transcription factor had eluded researchers because of their low abundance. Four years later, the GR would be the first transcription factor for a defined response element to be cloned, sequenced, and functionally identified.

A Rock and A Hard Place

A key question was whether the GR protein encoded by the receptor was sufficient, when expressed in a heterologous cell, to convey the hormonal message. Before the publication, a new postdoc, Vincent Giguere, began tinkering with the isolated GR, trying to address this question. The rate of development of any field is limited by the existing techniques and depends on the development of new ones. Vincent devised a revolutionary technique—the cotransfection assay that required two plasmids to be taken up in the same cell, the expression vector to be transcribed, the encoded protein to be functional and an inducible promoter linked to a chloramphenicol acetyltransferase reporter in the nucleus ready to flicker on (10, 12). With so many variables and unknowns, I was stunned and expressionless when it worked the very first time. Cotransfection was an easy, fast, and quantitative technique. It would become (and still remains) the dominant assay to characterize receptor function. It would also become the mainstay for drug discovery in the pharmaceutical industry. The development of this technique proved a great advantage because existing technology involved creating stable cell lines, a tedious process prone to integration artifacts that ultimately could not match the explosive pace of the field. Indeed, within 4 months Stan and Vincent had fully characterized 27 insertional mutants delineating the DBD, LBD, and two activation domains (12). The route to understanding the signaling mechanism now had a solid structural foundation. A serendipitous gift to my retroviral origins was the homology of the GR sequence to the v-erbA oncogene product of the avian erythroblastosis virus genome (13). With this discovery, erbA advanced to a candidate nuclear transcription factor potentially involved in a signal transduction pathway. Thus, while Stan concentrated on the GR, Cary began to delve into the erbA discovery. Within months of the GR publication, the human c-erbA gene was in hand (14). Unbeknownst to us, Bjorn Vennstrom, one of the first to characterize the avian erythroblastosis virus genome, had also isolated c-erbA and was searching for a function. Based on the low homology of the LBD region to the GR and ER, both groups deduced that the imaginary erbA ligand would be nonsteroidal.

The work of our two groups (15, 16), published in December of 1986, broadened the principles of the signal transduction pathway by demonstrating that thyroid and steroid hormone receptor signaling had a common evolutionary origin and provided an entree to understand how mutations within a receptor could activate it to an oncogene. Although we did not know it at the time, this work would also lead us to the concept of the corepressor. In the meantime, my student, Catherine Thompson, zeroed in on an erb-A-related gene and soon identified a second TR expressed at high levels in the central nervous system (17). Thus came into existence the and forms of the TR. Jeff Arriza, the third graduate student in the lab, purified a genomic fragment that had weakly hybridized to the GR resulting in the isolation of the human mineralocorticoid receptor (MR) (18). MR proved to have an at least 10-fold higher affinity for glucocorticoids than the GR itself and was further distinguished by its ability to bind and be activated by aldosterone. This enabled the development of GR- and MR-selective drugs such as the recent MR antagonist eplerenone. Thus, in a 2-yr time span our lab had progressed on three distinct, albeit related, receptor systems, and in doing so molecular biology and endocrinology were irrevocably linked. The field of molecular endocrinology (and coincidentally the eponymous journal) was born.

Ligands From Stone

I have often been asked how we could handle so many divergent systems. Indeed, from a medical perspective, these systems seem widely unrelated. Studies of ER, progesterone receptor, and androgen receptor (AR) fall under reproductive physiology, vitamin D under bone and mineral metabolism, with vitamin A part of nutritional science. Medical fields are naturally idiosyncratic because of the specialized knowledge required to conduct experiments. With my training as a molecular biologist, physiology was the complex output of genes and thus control of gene expression was the overriding problem. This conceptual approach had a great unifying effect because all receptors transduce their signaling through the gene. As an “outsider,” my goal was to exploit multiple receptor systems to seek general principles. This philosophical approach afforded us a freedom to redefine the signaling problem from the nucleus outward and thus even poorly characterized, even unknown, physiologic systems fell into the crosshairs of our molecular gun.

Vincent, while screening a testes Fig. 1. Models of Nuclear Receptor Structure Top, Original hand-shaped wire model (circa 1992) of the nuclear receptor DBD. Bottom, Schematic representation of the GR DBD. Conserved residues in zinc fingers, P-box and D-box are indicated isolated what would become the vitamin A or retinoic acid receptor (RAR) (19). Initially, Vincent thought he had isolated the AR, although this later proved not to be the case. By that stage, the lab had perfected a new technique—the domain swap—by which the GR DBD could be introduced into any receptor and confers on the chimeric protein the ability to activate a mouse mammary tumor virus reporter. This clever technique, independently developed in the Chambon lab, would prove to be essential. Effectively, the domain swap would enable us to screen for ligands without any knowledge of their physiologic function. Activation of a target gene was all that was needed! By creating this GR chimera, Vincent was able to screen the new receptor against a ligand cocktail including androgens, steroids, thyroid hormone, cholesterol, and the vitamin A metabolite retinoic acid. From the first assay, it was clear that he had isolated a high-affinity selective RAR that had no response to any other test ligand. Thus, without knowing any true direct target gene for retinoic acid, we were nonetheless able to isolate and characterize its receptor. Remarkably, Martin Petkovich in the Chambon lab isolated the same gene. Once again, this is an example where a new technique offered an entirely new approach to a problem. Both papers were published in the December 1987 issue of Nature (19, 20). With the combination of steroids, thyroid hormones, and vitamin A, the three elemental components of the nuclear receptor superfamily were in hand. By the time the RAR papers were published, Vincent with Na Yang, had already isolated two estrogen-related receptors termed ERR1 and 2 that would represent the first true orphan receptors in the evolving superfamily (21). A third receptor (ERR3) would be isolated 10 yr later (22). The three ERRs are distinguished by their ability to activate through ER response elements, but required no ligand. However, of potential major medical relevance, estrogen antagonists such as 4-hydroxy-tamoxifen silences ERR constitutive activity (23). The superfamily was growing exponentially, transforming into a new field, driven by a new breed of exceptional students and fellows attracted by the mechanics of transcription and its emerging link to physiology. For example, the RAR and TR offered an unprecedented look at understanding the action of vitamin A as a morphogen and the role of thyroxin in setting the basal metabolic rate of the body. We were a relatively small group, and our decision to work on multiple different receptor systems created a unique environment. Because there was so little overlap between projects, postdocs and students readily discussed all results, exchanged reagents and freely collaborated, resulting in a tremendous acceleration of progress. The high level of camaraderie was powered by the joie de vivre of the exciting discoveries and the ability of our family of students and postdocs to each adopt their own receptors. We all felt we were in a golden age and even more was to come.

In 1989, Jan Sap in Vennstrom’s group and Klaus Damm in our group demonstrated that the TR becomes oncogenic by mutation in the LBD (24, 25). Although we expected ligand-independent activation, it was clearly a constituitive repressor becoming the first example of a dominant-negative oncogene. The concept of the dominant-negative oncogene had been proposed one year earlier by Ira Herskowitz (26). This discovery changed our thinking on hormone action, and repression soon would be shown to be a common feature of receptor antagonists. David Mangelsdorf, who had arrived in the lab the year before was captivated by the glow of weakly hybridizing DNA bands and, in 1989, had amassed his own collection of orphan receptors, among which was the future retinoid X receptor (RXR) (27). In search for biological activity, a candidate ligand was found in lipid extracts from outdated human blood. However, the key test came from demonstrating that addition of all-trans retinoic acid to cultured cells would lead to its rapid metabolism coupled with the release of an inducing activity for RXR, which we termed retinoid X. David and his benchmate, Rich Heyman, began working on the chemistry of this inducer along with Gregor Eichele and Christine Thaller, then at Baylor College of Medicine (Houston, TX). This work led to the identification of 9-cis retinoic acid by our lab and a group at Hoffman LaRoche (Nutley, NJ) (28, 29). Like the retinal molecule in rhodopsin, 9-cis-retinoic acid represents the exploitation of retinoid isomerization by nature to control a key signaling pathway. More importantly, in the 39 yr since the discovery of aldosterone in 1953, this revelation would reawaken and reinvent the single most defining but dormant tool of endocrinology—ligand discovery. Indeed, the discovery that new receptors could lead to new ligands opened up an entirely new avenue of research. Like the puzzle of the structure of the benzene ring, which was solved in 1890 when Fredrick Kekule dreamed of a snake biting its own tail, the physiologic head of the “endocrine snake” and the molecular biology tail had come full circle. The era of reverse endocrinology was now upon us.

Response Elements: Deciphering The Scripts

One problem in addressing the downstream effects of our newly discovered receptors was that their response elements and target genes were by definition unknown. Kaz Umesono delved into this mystery and would produce a paradigm shift that would both solve the problem and further unify the field. With the view that the DBD functioned as a molecular receptor for its cognate hormone response element, meticulous mutational studies revealed two key DBD sequences, termed the P-box and D-box, that controlled the process (30).

The D-box was shown to direct dimerization, a feature previously thought to be unique to the LBD. One perplexing point was that the P-boxes of the nonsteroidal receptors were conserved, leading to the improbable prediction that many different receptors would recognize the same target sequence. By manual compilation and comparison of all known response elements, Kaz proposed a core hexamer— AGGTCA—as the prototypic common target sequence. By requiring the half-site to be an obligate hexamer an underlying pattern—the direct repeat—emerged. In the direct repeat paradigm, we proposed that half-site spacing, not sequence difference, was the key ingredient to distinguishing the response elements. The metric was referred to as the 3-4-5 rule (31). According to the rule, direct repeats of AGGTCA spaced by three nucleotides, would be a vitamin D response element (DR-3), the same repeat spaced by four nucleotides a thyroid hormone response element (DR-4), and the same repeat spaced by five nucleotides a vitamin A response element (DR-5). Eventually, all steps from 0–5 on the DR ladder would be filled (Fig. 2). The validity of this paradigm was ensured by a crystal structure solved in collaboration with Paul Sigler’s group at Yale (32). Indeed, of the remaining 40 nonsteroidal receptors, all but three can be demonstrated to have a preferred binding site within some component of the direct repeat ladder. Exceptions include SHP and DAX, which lack DBDs, and farnesoid X receptor (FXR) that binds to the ecdysone response element as a palindrome with zero spacing. Kaz’s insight, by drawing commonality from diversity, came to solve a problem that impacted on virtually every receptor. Remarkably, each new receptor in the superfamily could immediately be assigned a place on the ladder. The ladder also provided a simple means to conduct a ligand screening assay in absence of any knowledge of an endogenous target gene. Kaz’s ladder was a turbo charge for the field. The next major advance in the field was the discovery of the RXR heterodimer. Although we knew that retinoid and thyroid receptors required a nuclear competence factor for DNA binding, its identity was unknown. We tested RXR, but our initial experiments were flawed. Of the first four papers describing the discovery, that from Chambon’s lab was most elegant because they simply purified an activity to homogeneity to find RXR (33)! Rosenfeld was first to publish, and Ozato, Pfahl and Kliewer all concurred (34–37). Tony Oro and Pang Yao in our lab soon published that the ecdysone receptor functions as a heterodimer with ultraspiracle, the insect homolog of RXR (38, 39), revealing that the ancient origins of the heterodimer which arose before the divergence of vertebrates and invertebrates.

Reverse Endocrinology: Decoding Physiology

The orphan receptors would transform our view of endocrine physiology with unexpected links to toxicology, nutrition, cholesterol, and triglyceride metabolism as well as to a myriad of diseases including atherosclerosis, diabetes, and cancer. The three RXR isoforms formed the core with 14 heterodimer partners including the vitamin D receptor (VDR), TR/, and RAR//. The initial adopters of orphan receptors included Giguere, Mangelsdorf, Weinberger, Bruce Blumberg, Steve Kliewer, and Barry Forman. Barry unlocked the first secret to for peroxisome proliferator-activated receptor (PPAR) by identifying prostaglandin J2 (PGJ2) as a high-affinity ligand (40). The second step, in collaboration with Peter Tontonoz in Bruce Spiegleman’s lab, revealed that PGJ2 was adipogenic in cell lines and perhaps more importantly that the synthetic antidiabetic drug Troglitazone was a potent PPAR agonist (41). Similar work was conducted and published by Kliewer, who had now moved to Glaxo (42). By acquiring a ligand, a physiologic response, and a drug, PPAR was suddenly transported to the center of a physiologic cyclone that would spin into its own specialty field. Since 1995, more than 1000 papers (see PubMed) have been published on PPAR and its natural and synthetic ligands. This early work illuminated the molecular strategy of reverse endocrinology and the emerging importance of the orphan receptors in human disease and drug discovery. Cary returned to the lab for a sabbatical and, with Barry, demonstrated that FXR was responsive to farnesoids and other molecules in the mevalonate pathway. The findings by Mangelsdorf that liver X receptors (LXRs) bound oxysterols (43) and by Kliewer, Mangelsdorf, and Forman that FXR is a bile acid receptor (44–46) provided a whole new conceptual approach to cholesterol and triglyceride homeostasis. The steroid and xenobiotic receptors (SXR)/pregnane X receptor (PXR) (47–49) and the constituitive androstane receptor (CAR) (50) respond to xenobiotics to activate genes for P450 Fig. 2. Examples of Receptor Heterodimer Combinations that Fill the Direct Repeat (DR) Response Element Ladder from DR1 to DR5 Evans enzymes, conjugation and transport systems that detoxify drugs, foreign chemicals, and endogenous steroids. RXR and its associated heterodimeric partners quickly established a new branch of physiology, shedding its dependence on endocrine glands and allowing the body to decode signals from environmental toxins, dietary nutrients, and common metabolites of intermediary metabolism.

Continued…

ROCK OF AGES

The human body is, after all a living machine, a complex device that consumes and uses energy to sustain itself, defend against predators, and ultimately reproduce. One might reasonably ask, “If the superfamily acts through a common molecular template, can the family as a whole be viewed as a functional entity?” In other words, is there yet some overarching principle that we have yet to grasp. . . and might this imaginary principle lie at the heart of systems physiology? Simply stated, what led to the evolution of integrated physiology as the functional output of the superfamily? One obvious speculation is survival. To survive, all organisms must be able to acquire, absorb, distribute, store, and use energy. The receptors are exquisitely evolved to manage fuel—everything from dietary and endogenous fats (PPARs), cholesterol (LXR, FXR), sugar mobilization (GR), salt (MR), and calcium (VDR) balance and maintenance of basal metabolic rate (TR). Because only a fraction of the material we voluntarily place in our bodies is nutritional, the xenobiotic receptors (PXR, CAR) are specialized to defend against the innumerable toxins in our environment. Survival also means reproduction, which is controlled by the gonadal steroid receptors (progesterone receptor, ER, AR). However, fertility is dependent on nutritional status, indicating the presumptive communication between these two branches of the family. The third key component managed by the nuclear receptor family is inflammation. During viral, bacterial, or fungal infection, the inflammatory response defends the body while suppressing appetite, conserving fuel, and encouraging sleep (associated with cytokine release). However, if needed, even an ill body is capable of defending itself by releasing adrenal steroids, mobilizing massive amounts of fuel, and transiently suppressing inflammation. In fact, clinically, (with the exception of hormone replacement) glucocorticoids are only used as antiinflammatory agents. Other receptors including the RARs, LXRs, PPAR and , and vitamin D receptor protect against inflammation. Thus, nature evolved within the structure of the receptor the combined ability to manage energy and inflammation, indicating the important duality between these two systems. In aggregate, this commonality between distinct physiologic branches suggests that the superfamily might be approached as an intact functional dynamic entity.

Historically, endocrinologists and geneticists rarely saw eye to eye. As I have indicated in this perspective article, the disciplines have now become united in a new subject—transcriptional physiology. With this in mind, we might expect the existence of larger organizational principles that establish how the various evolutionary branches of the superfamily integrate to form whole body physiology. The existence of molecular rules governing the function and evolution of a megagenetic entity like the nuclear receptor superfamily, if correct, may be useful in understanding complex human disease and provide a conceptual basis to create more effective pharmacology. With so much accomplished in the last 20 yr (see Fig. 3), there are glimpses of clarity—enough to see the enormity and wonder of the problem and enough to know there is still a long and challenging voyage ahead. But who knows, the next breakthrough may only be a stone’s throw away.

http://press.endocrine.org/doi/pdf/10.1210/me.2005-0046

 

Pierre Chambon MD

Recipient of the Canada Gairdner International Award, 2010
“For the elucidation of fundamental mechanisms of transcription in animal cells and to the discovery of the nuclear receptor superfamily.”

Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch-Graffenstaden, France

Dr. Pierre Chambon is Honorary Professor at the College de France (Paris), and Emeritus Professor at the Faculté de Médecine of the Strasbourg University. He was the Founder and former Director of the IGBMC, and also the Founder and former Director of the Institut Clinique de la Souris (ICS/MCI), in Strasbourg.

Dr. Pierre Chambon is a world expert in the fields of gene structure, and transcriptional control of gene expression. During the last 25 years his studies on the structure and function of nuclear receptors has changed our concept of signal transduction and endocrinology. By cloning the estrogen and progesterone receptors, and discovering the retinoic acid receptor family, he markedly contributed to the discovery of the superfamily of nuclear receptors and to the elucidation of their universal mechanism of action that links transcription, physiology and pathology. Through extensive site-directed mutagenesis and genetic studies in the mouse, Pierre Chambon has unveiled the paramount importance of nuclear receptor signaling in embryonic development and homeostasis at the adult stage. The discoveries of Pierre Chambon have revolutionized the fields of development, endocrinology and metabolism, and their disorders, pointing to new tactics for drug discovery, and finding important applications in biotechnology and modern medicine.

These scientific achievements are logically inscribed in an uninterrupted series of discoveries made by Pierre Chambon over the last 45 years in the field of transcriptional control of gene expression in higher eukaryotes: discovery of PolyADPribose (1963), discovery of multiple RNA polymerases differently sensitive to a-amanitin (1969), contribution to elucidation of chromatin structure: the Nucleosome (1974), discovery of animal split genes (1977), discovery of enhancer elements (1981), discovery of multiple promoter elements and their cognate factors (1980-1993).

Pierre Chambon has received numerous international awards, including the 2004 Lasker Basic Medical Research Award for the discovery of the superfamily of nuclear hormone receptors and the elucidation of a unifying mechanism that regulates embryonic development and diverse metabolic pathways. He is a member of the French Académie des Sciences, and also a Foreign Member of the National Academy of Sciences (USA) and of the Royal Swedish Academy of Sciences. Pierre Chambon serves on a number of editorial boards, including Cell, and Molecular Cell. Pierre Chambon is author of more than 900 publications. He has been ranked fourth among most prominent life scientists for the 1983-2002 period.

An Interview with Pierre Chambon
2004 Albert Lasker Basic Medical Research Award
http://www.laskerfoundation.org/media/v_chambon.htm

Pierre Chambon, MD

​Honorary Professor at the Collège-de-France and Professor of Molecular Biology and Genetics, Institute for Advanced Study, University of Strasbourg; Group Leader, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch-Graffenstaden, Strasburg, France

A pioneer in the fields of gene structure and transcriptional control of gene expression, Dr. Chambon has fundamentally changed our understanding of signal transduction, which has led to revolutionary new tactics for drug discovery. His work elucidated how molecules that promote gene transcription are organized and regulated in eukaryotic organisms and, independently of Dr. Ronald Evans, he discovered in 1987 the retinoid receptor families, which led to the discovery and characterization of the superfamily of nuclear hormone receptors, including steroid and retinoid receptors.

Dr. Chambon’s previous research led to the discovery of PolyADPribose, multiple RNA polymerases differentially sensitive to α-amaniti, and has markedly contributed to the elucidation of the nucleosome and chromatin structure, as well as to the discovery of animal split genes, DNA sequences called enhancer elements, and multiple promoter elements and their cognate factors. These discoveries have greatly enhanced understanding of embryonic development and cell differentiation. To further studies of various nuclear receptors, Dr. Chambon has developed a method that allows in the mouse the generation of somatic mutations of any gene, at any time, and in any specific cell type, a tool valuable in generating mouse models of cancer.

In 1994, Dr. Chambon took on the role of founding a major research institute in France. As the first director of IGBMC, he built the institute to encompass hundreds of top researchers and multiple research programs funded by public agencies and private industry. In 2002, he founded and was the first director of the Institut Clinique de la Souris in Strasbourg. In these positions, he has succeeded in supporting and influencing a generation of scientists.

Career Highlights

​2010  Canada Gairdner International Award

2004  Albert Lasker Basic Medical Research Award

2003  Alfred P. Sloan, Jr., Prize, General Motors Cancer Foundation

1999  Louisa Gross Horwitz Prize, Columbia University

1998  Robert A. Welch Award in Chemistry

1991  Prix Louis-Jeantet de médecine, Fondation Louis-Jeantet

1990  Sir Hans Krebs Medal, Federation of European Biochemical Societies

1988  King Faisal International Prize for Science, King Faisal Foundation

1987  Harvey Prize, Technicon-Israel Institute of Technology

more…

 

Minireviews In This Series:

Thematic Minireview Series on Nuclear Receptors in Biology and Diseases

Sohaib Khan and Jerry B Lingrel

Steroid Receptor Coactivator (SRC) Family: Masters of Systems Biology

Brian York and Bert W. O’Malley

Estrogen Signaling via Estrogen Receptor β

Chunyan Zhao, Karin Dahlman-Wright, and Jan-Åke Gustafsson

Small Molecule Inhibitors as Probes for Estrogen and Androgen Receptor Action

David J. Shapiro, Chengjian Mao, and Milu T Cherian

Cellular Processing of the Glucocorticoid Receptor Gene and Protein: New Mechanisms for Generating Tissue Specific Actions of Glucocorticoids

Robert H Oakley and John A Cidlowski

Endogenous Ligands for Nuclear Receptors: Digging Deeper

Michael Schupp and Mitchell A. Lazar

 

 

 

Read Full Post »

Cancer and Nutrition

Writer and Curator: Larry H. Bernstein, MD, FCAP

The following discussions have been a topic of great interest and much controversy. In this discussion I shall not cover the topics related to Alternative and Complementary Medicine that is discussed elsewhere.  However, there is significant reason to explore the relationships of vitamin and micronutrient insufficiencies to cancer. The following nutritional subjects will be the focus of these discussions.

  1. Transthyretin (TTR)
  2. Vitamin A (retinoids and retinol) and retinol-binding protein (RBP)
  3. Vitamin C
  4. Vitamin D
  5. Magnesium (Mg++)

Cancer, homocysteine, Alzheimer’s Disease, and cardiovascular disease

1 Transthyretin

1.1 Plasma Transthyretin Indicates the Direction of both Nitrogen Balance and Retinoid Status in Health and Disease

Ingenbleek Yves1 and Bienvenu Jacques2,3,*

1Laboratory of Nutrition, Faculty of Pharmacy, University Louis Pasteur Strasbourg 1, France; 2Laboratory of Immunology, Hospices Civils de Lyon and 3INSERM U 851, University Claude Bernard Lyon 1, France

The Open Clinical Chemistry Journal, 2008;  1:1-12
Abstract: Whatever the nutritional status and the disease condition, the actual transthyretin (TTR) plasma level is determined by opposing influences between anabolic and catabolic alterations. Rising TTR values indicate that synthetic processes prevail over tissue breakdown with a nitrogen balance (NB) turning positive as a result of efficient nutritional support and / or anti-inflammatory therapy. Declining TTR values point to the failure of sustaining NB as an effect of maladjusted dietetic management and / or further worsening of the morbid condition. Serial measurement of TTR thus appears as a dynamic index defining the direction of NB in acute and chronic disorders, serving as a guide to alert the physician on the validity of his therapeutic strategy. The level of TTR production by the liver also works as a limiting factor for the cellular bioavailability of retinol and retinoid derivatives which play major roles in the brain ageing process. Optimal protein nutritional status, as assessed by TTR values within the normal range, prevents the occurrence of vascular and cerebral damages while maintaining the retinoid-mediated memory, cognitive and behavioral activities of elderly persons.

INTRODUCTION  Measurement of transthyretin (TTR, formerly called prealbumin) was proposed as nutritional marker in The Lancet in 1972 [1]. This proposal was largely disregarded by the scientific community during the decade following its publication. TTR testing is now the most utilized nutritional marker worldwide, having received the strong support of the Prealbumin Consensus Group [2].  A minority of workers, however, remain doubtful [3] or even reluctant [4] to adopt TTR as nutritional index, stressing the point that its synthesis is also influenced by inflammatory conditions [3,4] and by other extra-nutritional factors such as natural or synthetic corticosteroids [5] and androgens [6]. The aim of the present review paper is to clarify the complex relationships linking malnutrition and inflammation, throwing further insight into a nutrition domain of increasing public health.

EVOLUTION, STRUCTURE AND FUNCTIONS  TTR is a highly conserved protein in vertebrate species already secreted by the choroid plexus of reptiles 300 millions years ago and remaining confined within the cerebrospinal fluid (CSF) [10]. Synthesis and secretion of TTR by the liver evolved much later, about 100 millions years ago, in birds and eutherian mammals [11]. Production of TTR by the liver and by the choroid plexus is regulated separately [12]. The human TTR gene has been localized on the long arm of the chromosome 18q23 [13]. The nucleotide sequences of the entire TTR gene, including the 5′ (transcription initiating site) and the 3′ (untranslated site) flanking regions have been described [14,15]. The gene spans 6.9 kilobases (kb) and consists of 4 exons and 3 introns [14,15]. The hepatic TTR mRNA measures 0.7 kb encoding a pro-TTR-monomer undergoing a cleaving process to release the native TTR monomer [16]. Four identical subunits each 127 amino acids (AAs) length coalesce noncovalently to generate the fully mature nonglycosylated molecule whose molecular mass (MM) reaches 55 kDa [17]. Two binding sites for thyroid hormones are buried inside the central channel of the TTR heterodimer [18]. The secondary, tertiary and quarternary conformation structures of the TTR protein have been reported using 1.8 Å Fourier analysis [18]. One TTR  monomer binds to a small companion protein (21 kDa MM) to which a single retinol is bound (all-trans-retinol), hence its RBP denomination [19]. X-ray crystallographic studies have shown that RBP possesses an eight-stranded -barrel core that completely encapsulates the retinol molecule [20]. Under usual circumstances, RBP is almost entirely saturated with retinol, explaining that the 3 components of the retinol circulating complex (RCC) of 76 kDa MM has a close 1:1:1 stoichiometry [21]. Aggregation of TTR to holo-RBP occurs within the endoplasmic reticulum prior to extracellular RCC secretion [22].  The TTR protein was first discovered in human CSF in 1942 [23] and soon after in human serum. Human TTR transports about 20% of the intravascular pool of both thyroid hormones (Thyroxine [T4], triiodothyronine [T3]) and at least 90-95% of the retinol circulating pool. The term transthyretin was recommended by the International Nomenclature Committee [26] stressing the dual conveying role played by TTR in all eutherians.

The biological half-life of TTR is approximately 2 days [27] whereas that of holo-RBP (RBP + bound retinol) is half a day [28]. By contrast, apo-RBP devoid of its retinol ligand displays a significantly reduced half-life of 3.5 hr [28] and undergoes rapid glomerular leakage with subsequent tubular disintegration and recycling of its AA residues. It is therefore assumed that TTR plays an important role in the safeguard of the retinol pool. The catabolic site of TTR is mainly the liver, followed by muscle mass, skin and kidneys [29].  The TTR molecule displays microheterogeneity [30] and tissue deposits occur throughout the normal ageing processes [31]. In contrast, TTR is characterized by a very large genetic polymorphism affecting about 100 different point mutations [32], leading to misfolded forms of the protein and occurrence of amyloid disorders in several organs. The tetrameric TTR protein is recognized as a component of the normal pancreatic cell structure, preserving its integrity against the risk of apoptosis [40]. Finally, normal TTR production is required for the maturation of brain neural stem cells [41] and for the control of spatial reference memory performances [42].

SIGNIFICANCE OF TTR THROUGHOUT THE HUMAN LIFESPAN  Significant alterations in the levels of protein intakes by humans affect protein synthesis, turnover and breakdown and determine the outcome of total body N (TBN).  Anabolism occurs when the rate of AA incorporation into protein exceeds that of oxidative losses, yielding a positive NB. Catabolism is the result of protein breakdown prevailing over protein synthesis [43]. Increasing gestational age is accompanied by a slow and predictable rise in TTR values correlated with birth weight and proved useful in distinguishing between small, appropriate and large for gestational age infants [47,48]. Starting from birth until 100 years of age, our reference TTR values [54] are those collected in the monograph ” Serum Proteins in Clinical Medicine ” edited by the Foundation for Blood Research. The plasma TTR concentrations in healthy neonates are approximately two thirds those measured in healthy mothers and thereafter increase slowly until the onset of puberty without displaying sexual differences. The rate of protein synthesis similarly increases linearly during the prepubertal period [55], consistent with superimposable N accretion rates [56]. Human puberty is characterized by major hormonal and metabolic alterations leading to increased height velocity and weight gain [60]. The onset of puberty requires close interrelationships between the effects triggered by growth hormone and  insulin-like growth factors, by thyroid and steroid hormones, by insulin and sex hormones [60]. Whereas androgens strongly promote the development of muscle mass in males and lipolytic effects on visceral and subcutaneous fat, estrogens have minimal effect on the female musculature while stimulating the accrual of subcutaneous fat depots [60]. Body composition studies indicate prepubertal redistribution of FM and FFM with a significantly higher S-shaped elevation of FFM in male adolescents compared with the blunted curve recorded in teenaged girls [61,62]. TTR values manifest closely paralleled sex- and age-peculiarities in process of time that are best explained by the deeper androgenic impregnation of male subjects [6,43]. The musculature is by weight the main component of FFM, representing 37% of body mass [61].  In healthy adults, the sex-related difference in plasma TTR-RBP concentrations is maintained at plateau levels after sexual maturity [54,63]. Normal TTR plasma values are stabilized around 290-320 mg/L in males and around 250-280 mg/L in females [54,63]. Starting from the sixties, TTR concentrations progressively decline over time, disclosing a steeper slope in elderly men that reflects a relatively more rapid deterioration of their muscle mass [43]. As a result, the earlier TTR sexual difference disappears by about the age of 70 years [43]. This correlates with the age-dependent curvilinear drop of TBN, characterized by an accelerated decrease after 65 years [64]. Taken together, the plasma TTR evolutionary patterns reveal a parallelism with FFM so that TTR serves as an indicator of muscle mass. The data show that age and gender are significant co-variates of TTR which require separate blood reference values [54].

TTR AS INDEX OF PROTEIN DEPLETION / REPLETION STATES  There exists a long-lasting debate aimed at identifying the most effective protein sources, level of energy-yielding substrates and the proportion among these for the support of protein metabolism. Under usual conditions, glucose functions as the major energy substrate for protein synthesis. If the carbohydrate energy is lacking, glucose must be synthesized by gluconeogenesis, mainly from the conversion of endogenous or dietary protein [65]. This corresponds to a form of nutritional wastage which augments the cost of protein synthesis, as documented by an increased urinary excretion of urea. The above metabolic pattern stands in broad conformity with the concept that ” protein synthesis occurs in the flame of sugars ” [66].

FAO/WHO/UNU recommends for healthy adults the safe level of 0.75 g k-1 day-1 protein intake [67]. Although this amount of protein sustains normal growth and keeps unmodified the concentration of most biological parameters, such intake appears to be marginally inadequate to maintain the metabolic reserve capacities that are required to mount optimal responses to stress [68]. Studies have disclosed that TTR plasma level and pool size remain unaltered because its synthetic and catabolic rates are both downregulated concomitantly [69]. Changes occurring during prolonged starvation causes the N balance to turn negative despite efforts to minimize protein catabolism [70]. There is a direct correlation between the rate of liver protein synthesis and intrahepatic concentrations of individual free AAs [71]. It is likely that the dietary limitation of some AAs such as tryptophan [72] or leucine [73] could specifically exert inhibitory effects on the transcriptional [74] or translational [75] regulation of protein synthesis. Consequently, protein depletion causes a decrease in TTR mRNA [72,74,76].

Transcription of the TTR gene in the liver is directed by CCAAT/enhancer binding protein (C/EBP) bound to nuclear factor 1 (NF1) [74]. Multiple hepatocyte nuclear factors (HNFs) function in the regulation of TTR gene expression [77]. It has been recently shown that one of them (HNF-4) plays prominent roles before and after injury [78]. The drop of liver TTR mRNA levels to about half as an effect of protein deprivation [74] is accompanied by a corresponding diminished secretion of mature TTR molecules in the bloodstream.

The rapidly turning over TTR protein is exquisitely sensitive to any change in protein and/or energy supply, being clearly situated on the cutting edge of the equipoise. This is documented in preterm infants in whom AA supply is responsible for maintaining normal protein synthesis which may be somewhat modulated by fluctuations in energy intake [79]. In the declared stage of protein malnutrition, the serial measurement of TTR may serve to grade the severity of the disease spectrum, from mild [90] to severe [1] forms. Both metabolic and structural N compartments undergo exhausting processes as documented by the fall of nitrogenous compounds in the urine of protein-depleted subjects [91]. The relative dominance of urea over ammonia catabolites [92] reflects the more intense turnover rate of tissues belonging to the readily mobilizable N pool. Decreased TTR plasma values are indeed correlated with the involution of the gut mucosa [93] and with the extent of liver dysfunction, more pronounced in the kwashiorkor disease with massive hepatic steatosis than in marasmus with limited fatty liver infiltration [1]. The structural N compartment nevertheless participates in the loss of body protein reserves, consistent with the reduced urinary output of creatinine [91], 3-methylhistidine [94] and soluble hydroxyproline [95]. The resulting sarcopenia [96,97] and the concomitant depression of immune mechanisms [98,99] render an account of the higher morbidity / mortality rates affecting TBN-depleted patients identified by the lowest TTR and RBP plasma concentrations [100]. The mortality risk of malnourished children in Central Africa becomes likely when SA and TTR reach the threshold of 16 g /L and 65 mg /L, respectively [101].

During nutritional rehabilitation from protein malnutrition, the restoration of visceral proteins occurs at different rates depending on the type of protein and the size of its plasma pool. TTR and RBP recovery appears as the main result of increased production rates by the liver [102]. Most studies contend the view that the trajectory outlined for TTR correlates with the fluctuations of body N mass, especially during the anabolic phase of growth and clinical recovery from protein malnutrition. Using impedance parameters for assessing the N compartment still remaining in place in the stressed body of adults undergoing renal dialysis, nephrologists were able to demonstrate close relationships between TTR and phase angle, reactance and resistance values [105]. In elderly noninfected persons, FFM index measured by dual X-ray absorptiometry exhibits the highest correlation with TTR (r = 0.64) compared to RBP (r = 0.52) [106]

TTR AS NITROGEN INDEX IN INFLAMMATORY DISORDERS  Inflammatory disorders of any cause are initiated by activated leukocytes releasing a shower of cytokines working as autocrine, paracrine and endocrine molecules [107]. Cytokines regulate the overproduction of acute-phase proteins (APPs), notably that of CRP, 1-acid glycoprotein (AGP), fibrinogen, haptoglobin, 1-antitrypsin and antichymotrypsin [107]. APPs contribute in several ways to defense and repair mechanisms, being characterized by proper kinetic and functional properties [107]. Interleukin-6 (IL-6) is regarded as a key mediator governing both the acute and chronic inflammatory processes, as documented by data recorded on burn [108], sepsis [109] and AIDS [110] patients. IL-6-NF possesses a high degree of homology with C/EBP-NF1 and competes for the same DNA response element of the IL-6 gene [111]. IL-6-NF is not expressed under normal circumstances, explaining why APP concentrations are kept at baseline levels. In stressful conditions, IL-6-NF causes a dramatic surge in APP values [107,112] with a concomitant suppressed synthesis of TTR as demonstrated in animal [113] and clinical [114] experiments.  Under acute stressful conditions, protein turnover is strongly stimulated by augmented tissue breakdown (mainly in the muscle mass) and enhanced specific tissues synthesis (mainly in the liver and at the site of injury). Proteolysis releases AA residues which are preferentially incorporated into the hepatic precursor pool involved in the production of APPs [115,116]. The rate at which proteins are degraded generally exceeds the rate of AA mobilization for protein synthesis [117,118] yielding a net negative NB associated with an increased urinary output of urea and ammonia [119]. Creatininuria and 3-methylhistidinuria are significantly elevated and remain highly correlated (r = 0.97) attesting to the substantial participation of the skeletal musculature to the stress responses [117]. The gap between degradative and synthetic processes widens in proportion to the severity of injury, resulting in correspondingly increased urinary N catabolites [43]. Serious injury affecting otherwise healthy adults may trigger urinary N losses reaching 40 g/day or 250 g/week, which corresponds to about 15% of TBN [43]. In long-lasting debilitating disorders, the persisting negative NB may deplete the baseline body cell mass by about 45%, carrying ominous prognostic significance [120].

Inadequate nutritional management [122], multiple injuries, occurrence of severe sepsis and metabolic complications result in persistent proteolysis [124] and subnormal TTR concentrations [66]. The evolutionary patterns of urinary N output and of TTR thus appear as mirror images of each other, which supports the view that TTR might well reflect the depletion of TBN in both acute and chronic disease processes. Even in the most complex stressful conditions, the synthesis of visceral proteins is submitted to opposing anabolic or catabolic influences yielding ultimately TTR as an end-product reflecting the prevailing tendency. Whatever the nutritional and/or inflammatory causal factors, the actual TTR plasma level and its course in process of time indicates the exhaustion or restoration of the body N resources, hence its likely (in)ability to assume defense and repair mechanisms. The serial measurement of TTR appears as a dynamic tool pointing to the direction and magnitude of NB, predicting therefore the disease outcome. Hundreds of studies are reporting the clinical usefulness of TTR measurement.  TTR is recommended for the assessment and nutritional follow-up of a large panel of hospitalized patients in internal medicine settings [130,131], in general surgery [132,133] and intensive care units [134,135]. Low TTR values thus appear to nonspecifically reflect the extent of liver damage rather than its etiology. Liver N tissue only represents by weight a minor proportion of TBN but its intense turnover rate (10 to 20-fold more rapid than that of muscle tissue) [43] and its critical involvement in the orchestration of most major metabolic and immune pathways [145] explains why liver failure of any cause is usually associated with varying degrees of clinical malnutrition [142].

The nutritional management of kidney patients has met noticeable improvement along the past decades. Until the mid 1980s TTR was regarded as unreliable and discarded, leaving the way for the general use of SA in kidney studies. The turning point came in 1987 when a careful statistical analysis stated that TTR was the most representative marker within a large battery of currently measured parameters [149]. The most recent studies clearly incline towards the common use of TTR superseding that of SA [8, 151-155]. It has been confirmed, mainly in intensive care renal units, that the serial measurement of TTR works as a strong independent predictor of long-term survival, allowing identification of the patients in need of nutritional intervention [151,155] or at risk of reduced life expectancy [154, 155]. Using proportional hazards regression models, the relative risk of death was inversely related to TTR concentrations in 8,157 hemodialyzed patients [155]. TTR is currently measured as nutritional marker in tropical areas where bacterial, viral and parasitic diseases are still highly prevalent, usually in connection with defective immune and vitamin A status, including malaria[156], trypanosomiasis [157], schistosomiasis [158], measles[159], shigellosis [160], and AIDS patients exhibit declining TTR values as the morbid condition worsens [161].

In westernized societies, elderly persons constitute a growing population group. A substantial proportion of them may develop a syndrome of frailty characterized by weight loss, clumsy gait, impaired memory and sensorial aptitudes, poor physical, mental and social activities, depressive trends. Hallmarks of frailty combine progressive depletion of both structural and metabolic N compartments [162]. Sarcopenia and limitation of muscle strength are naturally involutive events of normal ageing which may nevertheless be accelerated by cytokine-induced underlying inflammatory disorders [163,164]. Depletion of visceral resources is substantiated by the shrinking of FFM and its partial replacement by FM, mainly in abdominal organs, and by the down-regulation of indices of growth and protein status [162]. Due to reduced tissue reserves and diminished efficiency of immune and repair mechanisms, any stressful condition affecting old age may trigger more severe clinical impact whereas healing processes require longer duration with erratical setbacks. As a result, protein malnutrition is a common finding in most elderly patients [165] with significantly increased morbidity and mortality rates [166,167].

Measurement of visceral protein status is proved useful throughout the entire ageing lifespan. A wide range of co-morbidities associated with defective protein nutritional status is described in aging persons who become more prone to develop pressure sores [163], osteoporosis [170], oral candidiasis [171] and nuclear cataract [172].  The isolation and purification of rat TTR [173] has allowed to set up animal models. In normal rats, TTR manifests highly significant correlations with nutrient intakes and with visceral and carcass N stores [174]. In tumor-bearing rats, the progressive exhaustion of body protein mass towards cachexia states is correlated with declining TTR values [175]. TTR is currently utilized as indicator of protein nutritional status in cancer patients [176,177]. TTR is held as the most powerful test overall for evaluating visceral protein status of children with solid tumors [178] and leukemias [179] both at the time of diagnosis and throughout chemotherapy. In bone marrow transplantation for malignancies, TTR accurately reflects at any point changes in the patient’s clinical status [180]. TTR has proved to be a useful marker of nutritional alterations with prognostic implications in large bowel cancer [181], bronchopulmonary carcinoid tumor [182], ovarian carcinoma [183] and bladder epithelioma [184]. Many oncologists have observed a rapid TTR fall 2 or 3 months prior to the patient’s death [181]. In cancer patients submitted to surgical intervention, most postoperative complications occurred in subjects with preoperative TTR  180 mg/L [185]. Two independent studies came to the same conclusion that a TTR threshold of 100 mg/L is indicative of extremely weak survival likelihood and that these terminally ill patients better deserve palliative care rather than aggressive therapeutic strategies [185,186].

The AGP/TTR couple is recommended in chronic inflammatory disorders, notably in several cancer types [192,193]. Working along the same lines is the prognostic inflammatory and nutritional index (PINI) [194] which is successfully applied on large cohorts of patients. TTR also participates in the development of screening formulas recently generated by innovative analytical tools such as surface-enhanced laser desorption/ionization (SELDI) or matrix-assisted laser desorption/ionization (MALDI) coupled with time of flight mass spectrometry (TOF-MS). The advent of these sophisticated and costly proteomic fingerprinting studies of serum or other biological fluids are nevertheless promising in that they tentatively strive to identify the early stages of several disease conditions such as hepatitis B [195], tuberculosis [196], Alzheimer’s disease [197] or neoplastic disorders [198]. These proteomic detecting systems usually combine classical APP reactants with some minor biological compounds scarcely measured in routine laboratory practice such as cathepsin D, hemopexin, neopterin or vitronectin. The fact that most, if not all, of these fingerprinting formulas embody TTR measurement indicates that there exists among workers a large consensus considering this carrier-protein as the most reliable indicator of protein depletion in morbid circumstances.

PROGRESS IN TTR RESEARCH : THE BRAIN AGEING PROCESS  Dementia, defined as significant memory impairment and loss of intellectual functions, is a common and devastating public health problem, affecting an estimated 2-4% individuals over the age of 65 years. Two distinct clinicopathological conditions are usually taken into consideration as causative factors: Alzheimer’s disease (AD), a chronic and continuously progressing illness for which the only widely accepted risk conditions are age and family history of the disease; and cerebral infarction, a brain deteriorating process evolving along episodic and repetitive bouts so as to generate a syndrome of multi-infarct dementia (MID) [199]. The rates of both AD and MID increase dramatically with age, leading to coexisting pathologies with intermingled symptomatology [200]. In support to this mixed cases concept are the report of equally increased blood-brain barrier permeability in both AD and MID patients [201] and the accumulation of amyloid -protein in the brain of MID subjects mimicking AD pathology [202]. There exists considerable overlap between AD and MID clinical symptoms, giving rise to a continuum of patients in whom pure AD and pure MID represent the two extreme poles [200].  The elevated homocysteine (Hcy) values found in AD patients [208,209] are reportedly associated with dementia [208,210].

The choroid plexus is the sole site of mammalian brain involved in TTR production [214]. Its synthesis rate by the choroid epithelium is estimated 25 to 100 times higher than that of the liver on a weight basis [215]. As a result, TTR is a major component of CSF, constituting 10 to 25 % of total ventricular proteins [216] conveying up to 80% of intrathecal thyroxine [217]. TTR thus constitutes an hormonal carrierprotein fulfilling important ontogenic and functional properties in mammalian nervous structures, a concept further corroborated by the observation of its increased CSF concentration during the neonatal period [218]. The data imply that choroidal TTR facilitates the uptake of thyroxine from the bloodstream, governing its transport and delivery to brain tissues following a kinetic model developed by Australian workers [219]. In comparison, CSF contains 10 to 100 times lower RBP and retinol concentrations than plasma whilst retinyl esters from dietary origin are virtually absent [220]. Although it has been reported that minute amounts of RBP could be produced within the neuraxis [221], the sizeable proportion of retinol molecules required for brain maturation utilizes the RCC transport system to reach the choroid plexus. The very high receptor binding affinity expressed by neural tissues for RBP molecules [222] is confined within the endothelial cells of the brain microvasculature and within the choroidal epithelial cells, the two primary sites of the mammalian blood-brain barrier [223]. The contrast between high RBP binding affinities and low intrathecal concentrations makes it likely that holo-RBP does not experience significant transchoroidal diffusion, strongly suggesting that its retinol ligand is released in free form and readily taken up by membrane or intracellular receptors of neural cells. The dual TTR production, plasma-derived and choroid-secreted, allows complementary stimulation of brain activities. Thyroid hormones and retinoids indeed function in concert through the mediation of common heterodimeric motifs bound to DNA response elements [224,225]. The data also imply that the provision of thyroid molecules within the CSF works as a relatively stable secretory process, poorly sensitive to extracerebral influences [12] as opposed to the delivery of retinoid molecules whose plasma concentrations are highly dependent on nutritional and/or inflammatory alterations [66]. This last statement is documented by mice experiments [226] and clinical investigations [227] showing that the level of TTR production by the liver operates as a limiting factor for retinol transport. Defective TTR synthesis determines the occurrence of secondary hyporetinolemia which nevertheless results from entirely different kinetic mechanisms in the two quoted studies [226,227].

In the TTR knock-out mice model, holo-RBP molecules are normally synthetized and secreted by the liver but undergo rapid kidney leakage in the absence of stabilizing TTR molecules [228]. Despite very low levels of plasma retinol (about 5 % of wild type), these targeted mutated animals remain healthy and fertile, implying that efficient compensatory mechanisms take place. No such increased urinary output of RBP molecules occurs in malnourished patients who develop in proportion to their declining protein status electroretinographic abnormalities and ocular lesions which are pathognomonic symptoms of vitamin A deficiency [229]. During nutritional rehabilitation of malnourished subjects, the 3 RCC components gradually return to normal ranges even without retinol or carotene supplementation, indicating that the retinyl esters normally sequestered in liver stellate cells mandatorily need diet-induced synthesis of new TTR molecules before undergoing retinol conversion and binding as holo-RBP ligand [227]. The prominent place occupied by TTR in defining distal retinoid bioavailability has been too long unrecognized despite the warning expressing that ” overlooking the crucial role of TTR in vitamin A-metabolism results in unachieved or even misleading conclusions ” [66].

Retinol is a precursor substrate that must undergo a two step oxidation procedure to release firstly retinal and thereafter the two active all-trans- and 13-cis-retinoic acids (RAs) [225,230]. The latter converting steps are regulated by retinaldehyde dehydrogenase (RALDH) enzymes whose major sites of expression are the olfactory bulb, the striatum and the hippocampus [231,232]. The intracellular activities exerted by retinoid compounds are mediated by a large variety of specific receptors among which are cellular-RBP (CRBP), cellular-RA-BP (CRABP), RA-nuclear receptors (RARs) and retinoid X receptors (RXRs), each composed of 3 subtypes [225,232]. Retinol is the rate-limiting determinant of the concentration of both RA derivatives [233], implying that any fluctuation in protein status might entail corresponding alterations in the cellular bioavailability of retinoid compounds, with all the more rapid effects as all-trans-RA has a short biological half-life of less than 1 hr [234]. Because protein malnutrition is a common finding in as much as 50 % of elderly AD and MID patients [235], many of them could well suffer permanent hyporetinolemia still accelerating the declining concentration of retinoid molecules observed over the course of normal ageing [231].  Dietary vitamin A is required to modulate early development of brain structure and differentiation [236] together with neuronal plasticity, memory functioning and neurotransmitter signaling during adulthood [237].

The normal decrease of brain retinoid molecules throughout the ageing process principally affects the above-described major sites of RA synthesis [238], a regressive alteration even more pronounced in AD patients [231]. In murine models, early depletion of retinoids causes deposition of amyloid -peptides [239], initiating the formation of Alzheimer plaques. In aged animals, cognitive and memory deficits are associated with down-regulation of the expression of retinoid receptors which may recover their full activities under RA supplementation [240]. Administration of RA similarly restores expression of proteins involved in the control of amyloidogenic pathways [241]. Along the same preventive line is the demonstration that retinol disaggregates preformed amyloid fibrils, more effectively than does RA [242].  Alternatively, TTR participates in the maintenance of memory and normal cognitive processes during ageing by acting on the retinoid signalling pathway as recently reported on TTR-null knock-out mice model [42,243]. Moreover, TTR may bind amyloid -peptide in vitro, preventing its transformation into amyloid neurofibrils [244].

Protein malnutrition, as assessed by diminished TTR plasma values, causes the elevation of Hcy concentrations [245]. There exists an inverse correlation between both TTR and Hcy parameters, explaining why malnourished elderly persons incur increasing risk of Hcy-depended thrombovascular complications [213]. The defective mechanism is situated at the level of cystathionine–synthase (CS), an enzyme governing the crossroad of remethylation and transsulfuration pathways [246]. Japanese workers have recently provided experimental validation of the metabolic anomaly, showing that rats given methionine (Met)-deprived nutriture manifest depressed CS activity with subsequent elevation of Hcy plasma levels [247]. Among all essential AAs consumed in human nutrition, Met is regarded as the most critically available because its withdrawal from the customary diet causes the deepest negative NB, being almost as great as when a protein-free regimen is ingested [248]. Met is implicated in a large spectrum of metabolic and enzyme activities and participates in the conformation of a large number of molecules of survival importance [213]. Due to the fact that plant products are relatively Met-deficient, vegan subjects are more exposed than omnivorous to develop hyperhomocysteinemia – related disorders [249]. Dietary protein restriction may promote supranormal Hcy concentrations which appears as the dark side of adaptive attempts developed by the malnourished and/or stressed body to preserve Met homeostasis.  Summing up, we assume that the low TTR concentrations reported in the blood [235] and CSF [250] of AD or MID patients result in impairment of their normal scavenging capacity [244] and in the excessive accumulation of Hcy in body fluids [245], hence causing direct harmful damage to the brain and cardiac vasculature. In addition, depressed TTR concentrations indirectly inhibit the multitude of retinoid-dependent cerebral functioning pathways [231,243] allowing the development of amyloidogenic processes [239]. The practical consequences of these findings imply that the correct assessment of nutritional status is recommended in all elderly patients. The mental and cognitive dysfunctions of old age that are not genetically programmed but result from varying energy, protein and vitamin-deficiencies may be substantially prevented and sometimes improved provided that appropriate nutritional measures are undertaken.

CONCLUDING REMARKS  In spite of classical criticisms [3,4], TTR is regarded as a robust and reliable indicator of protein nutritional. Taking into account the gender- and age-specificities, TTR appears as the sole plasma protein reflecting the fluctuations of TBN pools. The relationship linking alterations of TTR plasma levels with body N reserves are documented both in animal models [175] and in human subjects [105,106].  Uncomplicated malnutrition primarily affects the metabolic N pool, reducing protein syntheses and NB to levels compatible with survival, an adaptive response well identified by declining TTR values. In inflammatory disorders, both metabolic and structural N pools participate in varying proportions in the cytokine-induced responses of the stressed body, resulting in TBN shrinking and concomitant depression of TTR concentrations. Abatement of the stressful condition and/or efficient nutritional rehabilitation allows restoration to normal levels of both TBN pools and TTR values following parallel slopes. TTR thus appears as a dynamic index predicting the outcome of the disease. We attached more importance to the trend outlined by its serial appraisal than to any single measurement.  Whatever the causal factor, depletion of TBN reserves attenuates the body’s capacity to mount appropriate immune and repair mechanisms. A number of clinical investigations have advocated the level of plasma TTR as predictor of the length of hospital stay (LOS) and of mortality rate [252, 255]. Not surprisingly, unrecognized malnutrition entails longer LOS, increased number of complications and higher care costs whereas early detection and treatment of high risk patients significantly alleviate the financial burden of hospitalization while improving the prognostic outcome of the patients [252-256]. The last statement is documented by the first prospective and randomized survey showing that reduced morbidity and mortality rates are depending on protein N intake and correlated with rising TTR concentrations [257]. Providing elderly persons with optimal protein nutritional status in order to insure their protection against the risk of neurodeterioration is the last message released by the fascinating TTR plasma protein.

Points to consider:

  1. Protein energy malnutrition has an unlikely causal relationship to carcinogenesis. Perhaps the opposite is true. However, cancer has a relationship to protein energy malnutrition without any doubt.  PEM is the consequence of cachexia, whether caused by dietary insufficiency, inflammatory or cancer.
  2. Protein energy malnutrition leads to hyperhomocysteinemia, and by that means, the relationship of dietary insufficiency of methionine has a relationship to heart disease. This is the significant link between veganism and cardiovascular disease, whether voluntary or by unavailability of adequate source.

1.2 Downsizing of Lean Body Mass is a Key Determinant of Alzheimer’s Disease

Yves Ingenbleek, and Larry H. Bernstein
Journal of Alzheimer’s Disease 44 (2015) 745–754
http://dx.doi.org:/10.3233/JAD-141950

Lean body mass (LBM) encompasses all metabolically active organs distributed into visceral and structural tissue compartments and collecting the bulk of N and K stores of the human body. Transthyretin (TTR) is a plasma protein mainly secreted by the liver within a trimolecular TTR-RBP-retinol complex revealing from birth to old age strikingly similar evolutionary patterns with LBM in health and disease. TTR is also synthesized by the choroid plexus along distinct regulatory pathways. Chronic dietary methionine (Met) deprivation or cytokine-induced inflammatory disorders generates LBM downsizing following differentiated physiopathological processes. Met-restricted regimens downregulate the transsulfuration cascade causing upstream elevation of homocysteine (Hcy) safeguarding Met homeostasis and downstream drop of hydrogen sulfide (H2S) impairing anti-oxidative capacities. Elderly persons constitute a vulnerable population group exposed to increasing Hcy burden and declining H2S protection, notably in plant-eating communities or in the course of inflammatory illnesses. Appropriate correction of defective protein status and eradication of inflammatory processes may restore an appropriate LBM size allowing the hepatic production of the retinol circulating complex to resume, in contrast with the refractory choroidal TTR secretory process. As a result of improved health status, augmented concentrations of plasma-derived TTR and retinol may reach the cerebrospinal fluid and dismantle senile amyloid plaques, contributing to the prevention or the delay of the onset of neurodegenerative events in elderly subjects at risk of Alzheimer’s disease.

Transthyretin and Lean Body Mass in Stable and Stressed State

http://pharmaceuticalintelligence.com/2013/12/01/transthyretin-and-lean-body-mass-in-stable-and-stressed-state/

A Second Look at the Transthyretin Nutrition Inflammatory Conundrum

http://pharmaceuticalintelligence.com/2012/12/03/a-second-look-at-the-transthyretin-nutrition-inflammatory-conundrum/

Stabilizers that prevent transthyretin-mediated cardiomyocyte amyloidotic toxicity

http://pharmaceuticalintelligence.com/2013/12/02/stabilizers-that-prevent-transthyretin-mediated-cardiomyocyte-amyloidotic-toxicity/

Thyroid Function and Disorders

http://pharmaceuticalintelligence.com/2015/02/05/thyroid-function-and-disorders/

Proteomics, Metabolomics, Signaling Pathways, and Cell Regulation: a Compilation of Articles in the Journal http://pharmaceuticalintelligence.com

http://pharmaceuticalintelligence.com/2014/09/01/compilation-of-references-in-leaders-in-pharmaceutical-intelligence-about-proteomics-metabolomics-signaling-pathways-and-cell-regulation-2/

Malnutrition in India, high newborn death rate and stunting of children age under five years

http://pharmaceuticalintelligence.com/2014/07/15/malnutrition-in-india-high-newborn-death-rate-and-stunting-of-children-age-under-five-years/

Vegan Diet is Sulfur Deficient and Heart Unhealthy

http://pharmaceuticalintelligence.com/2013/11/17/vegan-diet-is-sulfur-deficient-and-heart-unhealthy/

How Methionine Imbalance with Sulfur-Insufficiency Leads to Hyperhomocysteinemia

http://pharmaceuticalintelligence.com/2013/04/04/sulfur-deficiency-leads_to_hyperhomocysteinemia/

Amyloidosis with Cardiomyopathy

http://pharmaceuticalintelligence.com/2013/03/31/amyloidosis-with-cardiomyopathy/

Advances in Separations Technology for the “OMICs” and Clarification of Therapeutic Targets

http://pharmaceuticalintelligence.com/2012/10/22/advances-in-separations-technology-for-the-omics-and-clarification-of-therapeutic-targets/

Sepsis, Multi-organ Dysfunction Syndrome, and Septic Shock: A Conundrum of Signaling Pathways Cascading Out of Control

http://pharmaceuticalintelligence.com/2012/10/13/sepsis-multi-organ-dysfunction-syndrome-and-septic-shock-a-conundrum-of-signaling-pathways-cascading-out-of-control/

Automated Inferential Diagnosis of SIRS, sepsis, septic shock

http://pharmaceuticalintelligence.com/2012/08/01/automated-inferential-diagnosis-of-sirs-sepsis-septic-shock/

 

 

1.3 Transthyretin Blocks Retinol Uptake and Cell Signaling by the Holo-Retinol-Binding Protein Receptor STRA6

Daniel C. Berry, Colleen M. Croniger, Norbert B. Ghyselinck, Noa Noya
Vitamin A is secreted from cellular stores and circulates in blood bound to retinol-binding protein (RBP). In turn, holo-RBP associates in plasma with transthyretin (TTR) to form a ternary RBP-retinol-TTR complex. It is believed that binding to TTR prevents the loss of RBP by filtration in the kidney. At target cells, holo-RBP is recognized by STRA6, a plasma membrane protein that serves a dual role: it mediates uptake of retinol from extracellular RBP into cells, and it functions as a cytokine receptor that, upon binding holo-RBP, triggers a JAK/STAT signaling cascade. We previously showed that STRA6-mediated signaling underlies the ability of RBP to induce insulin resistance. TTR blocks the ability of holo-RBP to associate with STRA6 and thereby effectively suppresses both STRA6-mediated retinol uptake and STRA6-initiated cell signaling. Consequently, TTR protects mice from RBP-induced insulin resistance, reflected by reduced phosphorylation of insulin receptor and glucose tolerance tests. The data indicate that STRA6 functions only under circumstances where the plasma RBP level exceeds that of TTR and demonstrate that, in addition to preventing the loss of RBP, TTR plays a central role in regulating holo-RBP/STRA6 signaling.

1.4 Transthyretin Amyloidosis

1.4.1 (Adapted from a Review in Amyloid: Int J Exp Clin Invest 3:44-56, 1996)

While it was expected that variations in clinical presentation (FAP-I, II, III, IV) were the result of heterogeneity in etiology or pathogenesis of the hereditary amyloidosis, it was not until the discovery by Costa, et al., in 1978 showing transthyretin as a constituent of the fibril deposits, that the biochemical basis of these syndromes could be pursued (Costa, et al., 1978).  This resulted in the discovery of the first variant form of transthyretin mutation reported in 1983.  In 1989 there were approximately 12 known mutations and in 2002 there are at least 90.  Over 80 of these mutations are associated with amyloidosis.  In addition, there is evidence that normal transthyretin may for amyloid especially in the heart and be the basis for senile cardiac amyloidosis (Westermark, 1990).

The transthyretin amyloidoses by definition are all associated with tissue deposits of fibrils having transthyretin as a major protein constituent.  While there are a number of other constituents of the amyloid deposits, including proteoglycan, amyloid P component, and various lipoproteins, it is transthyretin that is the essential ingredient in this type of amyloid.

It would appear that the signals for down regulating production of transthyretin (cytokines such as IL1 and IL6) are the same as those which cause the positive acute phase response of serum amyloid A and C reactive protein (Costa, et al., 1986).  The negative acute phase phenomenon of transthyretin is used by clinicians to monitor nutritional status of their patients.

Transthyretin is firmly entrenched in the phylogenetic evolution of vertebrate species being present in both birds and reptiles and its primary structure has been stable throughout evolution (Richardson, 1994).

While plasma transthyretin is predominantly synthesized by the adult liver, it is also synthesized by the choroids plexus of the brain and mRNA is also present in the retinal pigment epithelium, pituitary and pancreas19, 20 .  Choroid plexus synthesis would appear to be necessary for the thyroid hormone across the basement membrane into the cerebral spinal space.

The binding of RBP to transthyretin saves this small protein (21,000 daltons) from plasma clearance via filtration in the kidney.  However, when the complex gives up retinal, RBP dissociates from transthyretin and goes to meet its fate.  Transthyretin evidently can recirculate to bind more RBP-vitamin A.  Plasma residence time of transthyretin is approximately 20-24 hours, representing a plasma half-life of no more than 15 hours  (Benson, et al., 1996).  This is really very rapid turnover for a plasma protein, compared to plasma residence time of apolipoprotein AI which is 5 days, and that of albumin which is approximately 27 days (t ½ =19 days).

Most variants of transthyretin are not associated with amyloidosis.  Most variants of transthyretin are not associated with any postulated “hot spots” in the coding region.  The Ser6 variant is the only known polymorphism, prevalence of approximately 12% in the Caucasian population.  All the other mutations are present in less than 2% of the population, except in the restricted areas of Northern Sweden where greater than 2% of inhabitants have the Met30 gene and in African Americans, when considered as a group, where approximately 3% have a Val122Ile mutation.  One possible explanation of the large number of pathogenic mutations in transthyretin is that the amyloidosis is a delayed onset disease and, therefore, there is a lessened degree of selection against perpetuation of a pathogenic mutation.

Variations on the theme include the involvement of the vitreous of the eye in a number of the kindreds.  Approximately a third of transthyretin mutations are associated with vitreous deposits of amyloid; however, this finding is not uniform within families.  In different kindreds, a single mutation may have different presentations.  Most notably, Swedish patients with Met30 transthyretin have a high incidence of vitreous opacities with presentation at a fairly advanced age (58 years); whereas Portuguese patients have a lower incidence of vitreous opacities, but have presentation of neuropathy at an early age (mean 32 or 33 years).  Some transthyretin variants present as pure cardiomyopathy (e.g. Met111) (Frederikson, et al., 1962).   The Indiana/Swiss kindred (Ser84) has 100% incidence of cardiomyopathy (Benson and Dwulet, 1983) and this also appears to be true for the Appalachian kindred (Ala60) (Benson, et al., 1987).

Significant renal amyloidosis is less common than cardiac amyloidosis in most of the kindreds.  Recently attention has been directed toward kindreds having transthyretin amyloidosis with extensive leptomeningeal amyloid.  This is the hallmark of the Ohio kindred with oculoleptomeningeal amyloidosis (Gly30) (Goren, et al., 1980; Peterson, et al., 1997) and a recently reported kindred from Hungary (Gly18) in which the first clinical manifestation is dementia (Vidal, et al.,1996).  The His69 mutation has been associated with vitreous opacities alone (Zeldenrust, et al., 1994), but in another family causes oculoleptomeningeal amyloidosis.   Features of the disease in particular kindreds make familiarity with the different clinical expressions of the various transthyretin variants essential.

1.4.2 An insight to the conserved water mediated dynamics of catalytic His88 and its recognition to thyroxin and RBP binding residues in human transthyretin

Avik Banerjeea & Bishnu P. Mukhopadhyaya
http://dx.doi.org:/10.1080/07391102.2014.984632

Human transthyretin (hTTR) is a multifunctional protein involved in several amyloidogenic diseases. Besides transportation of thyroxin and vitamin-A, its role towards the catalysis of apolipoprotein-A1 and Aβ-peptide are also drawing interest. The role of water molecules in the catalytic mechanism is still unknown. Extensive analyses of 14 high-resolution X-ray structures of human transthyretin and MD simulation studies have revealed the presence of eight conserved hydrophilic centres near its catalytic zone which may be indispensable for the function, dynamics and stability of the protein. Three water molecules (W1, W2 and W3) form a cluster and play an important role in the recognition of the catalytic and RBP-binding residues. They also induce the reorganisation of the His88 for coupling with other catalytic residues (His90, Glu92). Another water molecule (W5) participate in inter-monomer recognition between the catalytic and thyroxin binding sites. The rest four water molecules (W6, W*, W# and W†) form a distorted tetrahedral cluster and impart stability to the catalytic core of hTTR. The conserved water mediated recognition dynamics of the different functional sites may provide some rational clues towards the understanding of the activity and mechanism of hTTR.

1.4.3 Amyloid Formation by Human Carboxypeptidase D Transthyretin-like Domain under Physiological Conditions*

Javier Garcia-Pardo, Ricardo Graña-Montes, Marc Fernandez-Mendez, et al.

Proteins can form amyloid aggregates from initially folded states. The transthyretin-like domain of human carboxypeptidase D forms amyloid aggregates without extensive unfolding. The monomeric transthyretin fold has an inherent propensity to aggregate due to the presence of preformed amyloidogenic structural elements. Generic aggregation from initially folded states would have a huge impact on cell proteostasis.

1.5 Evolutionary changes to transthyretin: evolution of transthyretin biosynthesis Samantha J. Richardson
FEBS Journal 276 (2009) 5342–53
http://dx.doi.org:/10.1111/j.1742-4658.2009.07244.x

Thyroid hormones are involved in growth and development, particularly of the brain. Thus, it is imperative that these hormones get from their site of synthesis to their sites of action throughout the body and the brain. This role is fulfilled by thyroid hormone distributor proteins. Of particular interest is transthyretin, which in mammals is synthesized in the liver, choroid plexus, meninges, retinal and ciliary pigment epithelia, visceral yolk sac, placenta, pancreas and intestines, whereas the other thyroid hormone distributor proteins are synthesized only in the liver. Transthyretin is synthesized by all classes of vertebrates; however, the tissue specificity of transthyretin gene expression varies widely between classes. This review summarizes what is currently known about the evolution of transthyretin synthesis in vertebrates and presents hypotheses regarding tissue-specific synthesis of transthyretin in each vertebrate class.

1.6  Distinctive binding and structural properties of piscine transthyretin

C Folli, N Pasquato, I Ramazzina, R Battistutta, G Zanotti, R Berni
FEBS Letters 555 (2003) 279-284
http://dx.doi.org:/10.1016/S0014-5793(03)01248-1

The thyroid hormone binding protein transthyretin (TTR) forms a macromolecular complex with the retinol-specific carrier retinol binding protein (RBP) in the blood of higher vertebrates. Piscine TTR is shown here to exhibit high binding affinity for L-thyroxine and negligible affinity for RBP. The 1.56 Ang resolution X-ray structure of sea bream TTR, compared with that of human TTR, reveals a high degree of conservation of the thyroid hormone binding sites. In contrast, some amino acid di¡erences in discrete regions of sea bream TTR appear to be responsible for the lack of protein-protein recognition, providing evidence for the crucial role played by a limited number of residues in the interaction between RBP and TTR. Overall, this study makes it possible to draw conclusions on evolutionary relationships for RBPs and TTRs of phylogenetically distant vertebrates.

1.7 Protein  Synthesis  at the Blood-Brain Barrier: The Major Proteins  Ecreted By Amphibian Choroid Plexus Is A Lipocalin

  1. Achen, PJ. Harms, T Thomas, SJ. Richardson, REH. Wettenhall, G Schreiber J Biol Chemistry Nov 1992; 267(32): 23167-70Among the proteins secreted by choroid plexus of vertebrates, one protein is much more  abundant than all others. In  mammals, birds, and reptiles  this protein is transthyretin, a tetramer of identical 15-kDa sub- units. In this study choroid plexus from frogs, tadpoles, and toads incubated in  vitro were found to synthesize and secrete one predominant protein. However, this consisted of one single 20-kDa polypeptide chain. It was expressed throughout  amphibian metamorphosis. Part of its amino acid sequence was determined and used for construction of oligonucleotides for polymerase chain reaction. The amplified DNA was used to screen a toad choroid plexus cDNA library. Full-length cDNA clones were isolated and sequenced. The derived amino acid sequence for the encoded protein was 183 amino acids long, including a 20-amino acid preseg- ment. The calculated molecular weight of the mature protein was 18,500. Sequence comparison with other proteins showed that the protein belonged to the lipocalin superfamily. Its expression was highest in choroid plexus, much lower in other brain areas, and absent from liver.  Since no transthyretin was detected in proteins secreted from amphibian choroid plexus, abundant synthesis and secretion of transthyretin in choroid plexus must have  evolved only after the stage of the amphibians.

2 Vitamin A

2.1 Retinoic acid pathways and cancer

2.1.1 Vitamin A, Cancer Treatment and Prevention: The New Role of Cellular Retinol Binding Proteins

Elena Doldo,Gaetana Costanza,Sara Agostinelli,Chiara Tarquini, et al.
BioMed Research International 2015; Article ID 624627, 14 pages
http://dx.doi.org/10.1155/2015/624627

Retinol and vitamin A derivatives influence cell differentiation, proliferation, and apoptosis and play an important physiologic role in a wide range of biological processes. Retinol is obtained from foods of animal origin. Retinol derivatives are fundamental for vision, while retinoic acid is essential for skin and bone growth. Intracellular retinoid bioavailability is regulated by the presence of specific cytoplasmic retinol and retinoic acid binding proteins (CRBPs and CRABPs). CRBP-1, the most diffuse CRBP isoform, is a small 15KD acytosolic protein widely expressed and evolutionarily conserved in many tissues. CRBP-1 acts as chaperone and regulates the uptake, subsequent esterification, and bioavailability of retinol. CRBP-1 plays a major role in wound healing and arterial tissue remodeling processes. In the last years, the role of CRBP-1-related retinoid signaling during cancer progression became object of several studies. CRBP-1 downregulation associates with a more malignant phenotype in breast, ovarian, and nasopharyngeal cancers.Reexpression of CRBP-1 increased retinol sensitivity and reduced viability of ovarian cancer cells in vitro. Further studies are needed to explore new therapeutic strategies aimed at restoring CRBP-1-mediated intracellular retinol trafficking and the meaning of CRBP-1 expression in cancer patients’ screening for a more personalized and efficacy retinoid therapy.

Metabolism of Retinol and Its Derivatives. Vitamin A can be acquired from the diet either as preformed vitamin A (primarily as retinyl ester, retinol, and in much smaller amount as retinoic acid) or provitamin A carotenoids (Figure1). Dietary retinyl esters are converted to retinol within the lumen of the small intestine or the intestinal mucosa and then reesterified to form retinyl ester (RE) within the enterocyte [1]. Provitamin A carotenoids, absorbed by the mucosal cells, are converted first to retinaldehyde and then to retinol [1]. After secretion of the nascent chylomicrons into the lymphatic system, the bulk of dietary vitamin A is taken up by hepatocytes and hydrolyzed again.The free retinol binds the epididymal retinoic acid binding protein (ERABP) and the retinol binding protein (RBP) [2] and into plasma transthyretin. Free retinol can be transferred to hepatic stellate cells for storage. Hepatocytes and hepatic stellate cells are very rich in retinyl ester hydrolases and in cellular retinol binding protein type 1 (CRBP-1). CRBP-1 is necessary to solubilize retinol in the aqueous environment of the cell [1].

Intracellular Trafficking of Retinoids. A cell-surface receptor named stimulated by retinoic acid 6 (STRA6) mediates vitamin A uptake from RBP [3]. Intracellular retinoid bioavailability is regulated by the presence of specific cytoplasmic retinol and retinoic acid binding proteins, CRBPs and CRABPs (Figure2). In the cytoplasm vitamin A and derivatives are bound to cytoplasmic proteins: cellular retinol binding proteins (CRBPs) which comprised four isoforms, CRBP-1 and CRBP-2 and CRBP-3 and CRBP-4. CRBP-1, are the most represented isoform in many tissues. Cellular retinoic acid binding proteins (CRABPs) comprised two isoforms, CRABP-1 and CRABP-2. CRBPs specifically bind retinol, while CRABPs and well-characterized members of the fatty acid binding proteins (FABPs) bind retinoic acid (RA). These proteins control the availability of ligands and determine the physiological response of cells and tissues to vitamin A [4]. Cellular retinoic acid binding proteins may regulate the interactions between retinoic acids and their nuclear receptors by regulating the concentrationof present retinoic acids [5]. Retinoids can activate gene expression by specific nuclear retinoid acid receptors. Two distinct classes of nuclear proteins, the retinoic acid receptors (RARs), and the retinoid X receptors (RXRs) have been identified. Each class consists of 𝛼, 𝛽,and 𝛾 subtypes. RARs and RXRs form either homodimers or heterodimers and function as transacting nuclear transcriptional factors [6]. RAR can be activated by both all-trans and 9-cis RA, whereas RXR is only activated by 9-cis-RA.

2.1.2 Retinoids, retinoic acid receptors, and cancer.

Tang XH1, Gudas LJ.
Annu Rev Pathol. 2011; 6:345-64
http://dx.doi.org:/10.1146/annurev-pathol-011110-130303

Retinoids (i.e., vitamin A, all-trans retinoic acid, and related signaling molecules) induce the differentiation of various types of stem cells. Nuclear retinoic acid receptors mediate most but not all of the effects of retinoids. Retinoid signaling is often compromised early in carcinogenesis, which suggests that a reduction in retinoid signaling may be required for tumor development. Retinoids interact with other signaling pathways, including estrogen signaling in breast cancer. Retinoids are used to treat cancer, in part because of their ability to induce differentiation and arrest proliferation. Delivery of retinoids to patients is challenging because of the rapid metabolism of some retinoids and because epigenetic changes can render cells retinoid resistant. Successful cancer therapy with retinoids is likely to require combination therapy with drugs that regulate the epigenome, such as DNA methyltransferase and histone deacetylase inhibitors, as well as classical chemotherapeutic agents. Thus, retinoid research benefits both cancer prevention and cancer treatment.
2.1.3 Molecular pathways: current role and future directions of the retinoic acid pathway in cancer prevention and treatment.

Connolly RM1Nguyen NKSukumar S.
Clin Cancer Res. 2013 Apr 1; 19(7):1651-9
http://dx.doi.org:/10.1158/1078-0432.CCR-12-3175

Retinoids and their naturally metabolized and synthetic products (e.g., all-trans retinoic acid, 13-cis retinoic acid, bexarotene) induce differentiation in various cell types. Retinoids exert their actions mainly through binding to the nuclear retinoic acid receptors (α, β, γ), which are transcriptional and homeostatic regulators with functions that are often compromised early in neoplastic transformation. The retinoids have been investigated extensively for their use in cancer prevention and treatment. Success has been achieved with their use in the treatment of subtypes of leukemia harboring chromosomal translocations. Promising results have been observed in the breast cancer prevention setting, where fenretinide prevention trials have provided a strong rationale for further investigation in young women at high risk for breast cancer. Ongoing phase III randomized trials investigating retinoids in combination with chemotherapy in non-small cell lung cancer aim to definitively characterize the role of retinoids in this tumor type. The limited treatment success observed to date in the prevention and treatment of solid tumors may relate to the frequent epigenetic silencing of RARβ. Robust evaluation of RARβ and downstream genes may permit optimized use of retinoids in the solid tumor arena.

Vitamin A is derived from animal and plant food sources and has critical functions in many aspects of human biology. Its natural derivatives and metabolized products (retinoids) such as β-carotene, retinol, retinal, isotetrinoin, all-trans retinoic acid (ATRA), 9-cis retinoic acid, and 13-cis retinoic acid have important roles in cell differentiation, growth, and apoptosis (1). Synthetic retinoids are also available and include bexarotene and fenretinide. In clinical practice, retinoids have a wide range of dermatologic indications including for psoriasis, acneiform, and keratinization disorders (2). Systemic retinoids are approved by the U.S. Food and Drug Administration (FDA) for the treatment of cutaneous T-cell lymphoma (3) and acute promyelocytic leukemia (APL; refs. 4, 5). However, the chemopreventive and therapeutic effects of retinoids in solid tumors remain controversial. Therefore, an overview of the research to date and future directions in this area is the focus of this review.

Retinoic acid and the retinoic acid receptor pathway

Retinoic acids (RA) exert their functions through their specific receptors. The 2 distinct classes of receptors are retinoic acid receptors (RAR) and retinoic X receptors (RXR). Each class contains 3 different subtypes—α, β, and γ (6). ATRA and fenretinide can bind specifically to RARS, 13-cis RA and bexarotene only to RXRS, and 9-cis RA to RARS or RXRS (refs. 1, 5; Table 1). The expression of these receptors is regulated by the receptors themselves, other nuclear receptors such as ERα, or by other subtypes in the same family (5, 7). Upon the binding of ligands, RARs and RXRs form heterodimers and function as ligand-dependent transcription factors to activate their downstream effectors by binding to the retinoic acid response elements (RARE) located in the 5′-region of RA downstream genes (5). The above model of RAR or RXR function via binding to RARE is considered the RA classical or genomic pathway. Activation of the classical pathway will trigger cell differentiation, cell arrest, and eventual apoptosis (8).

Table 1. Select clinical trials evaluating retinoids in solid tumors

Retinoid Other names Target Clinical trial setting
ATRA Tretinoin RAR Advanced NSCLC Phase II randomized (n = 107)
13-cis RA Isotretinoin Roaccutane Accutane RXR Primary prevention: H+N cancer
Advanced solid tumorsPhase I (n = 13)
Metastatic breast cancer Phase II randomized (n = 99)
9-cis RA Alitretinoin RAR RXR Metastatic breast cancerPhase I (n = 12)
Fenretinide 4-OH Phenylretinamide RAR Primary prevention: women at high risk of breast cancer Randomized double-blind 2 × 2 design (n = 235)
Secondary prevention: early breast cancerPhase III randomized (n = 2,867)
Bexarotene RXR Chemotherapy-naïve advanced NSCLC Phase III randomized (n = 623)
Metastatic  breast cancer Phase II single arm (n = 148)

The function of RA and its receptors involves not only the classical pathway but also multiple other important pathways. RAs have been shown to regulate NF-κB (9), IFN-γ (10), TGF-β (11), VEGF (12), mitogen-activated protein kinase (MAPK; ref. 13), and chromatin remodeling (14). Furthermore, RARs and RXRs can form heterodimers with other types of receptors, including the estrogen receptor-α (ERα; refs. 7, 15), AP-1 receptor (16), peroxisome proliferator-activated receptor (PPAR; ref. 17), liver X receptors (LXR; refs. 18, 19), and vitamin D receptor (VDR; ref. 20; Fig. 1). When RARs/RXRs heterodimerize with these receptors, they are involved in regulating their partner receptor’s pathways, referred to as nonclassical or nongenomic pathways (5). Interestingly, these pathways often regulate processes that have functions opposite to the classical pathway. For example, a study has shown that RA activation of the PPARβ/δ pathway resulted in upregulation of prosurvival genes (17), contrary to the known differentiation function of RARs and RXRs in response to RA. The function of RAs, which involves nongenomic pathways, may provide opportunities for cancer cells to develop resistance to RA treatment, discussed later in this review. Another important function of RARA is the regulation of stem cell differentiation (11). RAs target stem cells via both genomic and nongenomic pathways such as the Notch pathway and inflammation (10, 11). In summary, RAs and their receptors play important roles as regulators of critical processes in cells.

RARs and their action

RARs and their action

The RARs and their action. In a series of enzymatic steps, vitamin A (retinol) is metabolized through the oxidizing action of retinaldehyde (RDH) to retinal, and by retinaldehyde dehydrogenase (RALDH), to RA. RA has 3 different isomers: all-trans, 9-cis, and 13-cis RA. RA is transported to the nucleus by the protein cellular RA–binding protein (CRABP) and delivered to the RARα. RARα heterodimerizes with and binds to RARE present most often in gene promoters. In the classical pathway of RA action, RA binds to dimers of RARα and RXRs (α, β, or γ) to induce expression of its downstream target genes, including RARβ. Upon activation, RARβ can regulate its own expression and that of its downstream genes, the function of which is mainly to inhibit cell growth. Alternatively, RA can be bound and transported to the nucleus by other factors such as FABP5. This delivers RA to other nonclassical receptors such as PPARβ/δ and ERα which activate nongenomic pathways such as PDK-1/Akt or the ERα pathway. Contrary to the differentiation functions attributed to the classical pathway, the nongenomic pathways exert strong antiapoptotic and proliferative effects on cancer cells. It is believed that the classical and nongenomic pathways are controlled by the relative abundance of their own ligands. RA has a stronger affinity for RARs than for the other receptors, and the classical pathway plays a dominant role over the nongenomic pathways. Thus, if RA is present with other ligands such as estrogen, signaling through the classical pathway is preferred to result in cell differentiation and growth inhibition.

http://clincancerres.aacrjournals.org/content/19/7/1651/F1.small.gif

Retinoids and cancer

The retinoids have been investigated extensively for the prevention and treatment of cancer, predominantly because of their ability to induce cellular differentiation and arrest proliferation. RA-regulated tumor suppressor genes, when expressed, can inhibit tumor growth (21). Among the 3 RARs, RARβ has been well known for its tumor-suppressive effects in epithelial cells (5822). Exogenous expression of the RARβ gene can cause RA-dependent and -independent apoptosis and growth arrest (23). RARβ-induced growth arrest and apoptosis is mediated through RARα (24). As RA ligand-bound RARα binds to the RARE on the RARβ promoter, multiple activator proteins assemble at the site and result in the upregulation of the RARβ gene (5). The expression of RARβ results in the transactivation and expression of a number of its target genes that mediate cell differentiation and death (5, 68). The ability of ATRA to initiate differentiation of promyelocytic leukemic cells to granulocytes is the basis of the dramatic success of retinoic acid therapy for acute promyelocytic leukemia harboring the RAR/PML translocation (4) and confirms the important role of RARβ in tumor growth inhibition. It is also becoming increasingly clear that RARβ expression is lost early in carcinogenesis or is epigenetically silenced (25) in many solid tumors, providing an opportunity for novel treatment strategies to be investigated using retinoids together with epigenetic modifiers that promote reexpression of silenced genes, described further below.

The retinoids have an established role in the treatment of certain hematologic malignancies, with FDA approval for use in cutaneous T-cell lymphoma and APL. Bexarotene (an RXR-selective retinoid or rexinoid) is associated with an overall response rate of approximately 50% in patients with refractory advanced-stage mycosis fungoides, a cutaneous T-cell lymphoma (3). ATRA, a synthetic retinoid, exhibited improvements in disease-free and overall survival when compared with chemotherapy alone in APL, with long-term remissions occurring in almost 70% of cases (4). The success of retinoids in treating this disease relates to the underlying chromosomal translocation and production of the PML/RARα fusion protein and the ability of retinoids to induce differentiation and inhibition of cell growth in this setting (26, 27). Clinical trials investigating the role of retinoids in the prevention and treatment of solid tumors will now be outlined with a focus on cancers of the upper aerodigestive tract (oropharyngeal and lung) and breast (Table 1).

2.1.4 Retinoid Pathway and Cancer Therapeutics

Nathan Bushue and Yu-Jui Yvonne Wan
Adv Drug Deliv Rev. 2010 Oct 30;  62(13): 1285–1298.
http://dx.doi.org/10.1016%2Fj.addr.2010.07.003

The retinoids are a class of compounds that are structurally related to vitamin A. Retinoic acid, which is the active metabolite of retinol, regulates a wide range of biological processes including development, differentiation, proliferation, and apoptosis. Retinoids exert their effects through a variety of binding proteins including cellular retinol binding protein (CRBP), retinol-binding proteins (RBP), cellular retinoic acid-binding protein (CRABP), and nuclear receptors i.e. retinoic acid receptor (RAR) and retinoid × receptor (RXR). Because of the pleiotropic effects of retinoids, understanding the function of these binding proteins and nuclear receptors assists us in developing compounds that have specific effects. This review summarizes our current understanding of how retinoids are processed and act with the emphasis on the application of retinoids in cancer treatment and prevention.

Vitamin A and its derivatives (retinoids) exert a wide range of effects on embryonic development, cell growth, differentiation, and apoptosis. Vitamin A has been used as a treatment for thousands of years. The Egyptian papyruses Kahun 1 (ca. 1825 B.C.) and Ebers (ca. 1500 B.C.) described how the liver was used to cure eye diseases such as night blindness. Greek scholar Hippocrates (460-327 B.C.) described in the second book of “Prognostics” a method for curing night blindness: “raw beef liver, as large as possible, soaked in honey, to be taken once or twice by mouth.” Chinese medicine used pigs’ liver as a remedy for night blindness, as described by Sun-szu-mo (7th century A.D.) in his “1000 Golden Remedies”. Given that the liver is where the body stores excess vitamin A, the liver represents the best source of vitamin A available for treatment in the pre-pharmaceutical world.

The effect of vitamin A on growth was first described in a mouse experiment done by G. Lunin (1881) [2], in which one group of mice was fed pure casein, fat, sucrose, minerals, and water, and another group was fed whole dried milk. The milk-fed group was healthy and grew normally, while the other group was sick and ultimately died. Thus, something in milk was essential for survival. Elmer McCollum at University of Wisconsin-Madison as well as Lafayette Mendel and Thomas Burr Osborne at Yale University independently discovered vitamin A. McCollum began his study in 1907 by feeding cows hay with wheat, oats, or yellow maize.

Wheat-fed cows did not thrive, became blind and gave birth to dead calves prematurely. Oat-fed cows fared somewhat better, but the yellow maize-fed cows were in excellent condition, produced vigorous calves, and had no miscarriages. McCollum postulated that performing the same nutritional study using small animals, such as rodents, which require less food, provide faster reproduction and experimental outcome. Using rats, he found a diet of pure protein, pure milk sugar, minerals, and lard (or olive oil) inhibited growth, while addition of butterfat or an ether extract of egg yolk to the diet restored health. Thinking that he had found a fat-soluble factor that promoted growth in rats, he saponified butterfat, extracted the unsaponifiable mixture into ether, and added the extract to oliveoil and that extract could support growth. This essential component to support growth and development was named “fat-soluble factor A,” and later renamed vitamin A [1].

There are over 4,000 natural and synthetic molecules structurally and/or functionally related to vitamin A. Vitamin A cannot be synthesized by any animal species and is only obtained through diet in the form of retinol, retinyl ester, or β-carotene (Figure 1). Ingested vitamin A is stored as retinyl esters in hepatic stellate cells. Retinol is reversibly oxidized by retinol dehydrogenases to yield retinal. Subsequently, retinal may be irreversibly oxidized to all-trans retinoic acid (all-trans RA) by retinal dehydrogenases and further oxidized by cytochrome P450 enzymes (mainly CYP26) in hepatic tissue. Retinol has six biologically active isoforms that include all-trans, 11-cis, 13-cis, 9, 13-di-cis, 9-cis, and 11, 13-di-cis, with all-trans being the predominant physiological form. Endogenous retinoids with biological activity include all-trans RA, 9-cis RA, 11-cis retinaldehyde, 3,4-didehydro RA, and perhaps 14-hydroxy-4, 14-retro retinol, 4-oxo RA, and 4-oxo retinol [35]. All-trans RA isomerizes under experimental and physiological conditions. Different isomers activate different receptors and thus lead to different biological effects. RAs designed to be receptor specific can improve efficacy and avoid unwanted side effects. Retinoids that specifically bind to RXR are called rexinoids and have been effective in cancer treatment. Retinoids are comprised of three units: a bulky hydrophobic region, a linker unit, and a polar terminus, which is usually a carboxylic acid. Modification of each unit has generated many more compounds. Please refer to recent reviews [68].

2.1.4.1 Retinoid Pathway 

Retinoid Pathway  nihms229611f1

Retinoid Pathway nihms229611f1

http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2991380/bin/nihms229611f1.jpg

Retinoids absorbed from food are converted to retinol and bound to CRBP in the intestine. Then, retinol is converted to retinyl esters and enters into blood circulation. The liver up takes retinyl esters, which are converted to retinol-RBP complex in the hepatocyte. In the serum, the retinol-RBP complex is bound to transthyretin (TTR) in a 1:1 ratio to prevent elimination by the kidney and to ensure retinol is delivered to the target cell. The uptake of retinol by the target cell is mediated by a trans-membrane protein named “stimulated by retinoic acid 6” (STRA6), which is a RBP receptor. In the target cell, retinol either binds to CRBP or is oxidized to retinaldehyde by retinol dehydrogenase (RDH) in a reversible reaction. Then, retinaldehyde can be oxidized by retinaldehyde dehydrogenase (RALDH) to RA. In the target cell, RA either binds to CRABP or enters the nucleus and binds to nuclear receptors to regulate gene transcription. Alternatively, RA can mediate via nongenomic mechanism and regulate cellular function. Hepatocytes not only process retinoids, but also are the target cells. In addition, hepatocytes located next to the storage site (stellate cell). Thus, retinoid-mediated signaling must have a profound effect in regulating hepatocyte function and phenotype [36190191].

2.1.4.2 Retinoid Binding Proteins

There are various types of retinoid-binding proteins, which locate in intracellular and extracellular compartments and associate with isomeric forms of retinoids. Hence, retinoids are either associated with cellular membranes or bound to a specific retinoid binding protein. These binding proteins along with nuclear receptors mediate the action of retinoids. Their interactions are summarized in figure 1. Retinoid-binding proteins solubilize and stabilize retinoids in aqueous spaces. In addition to this general role, specific retinoid-binding proteins have distinct functions in regulating transport and metabolism of specific retinoids. For example, the parent vitamin A molecule, all-trans retinol, circulates in blood bound to serum retinol binding protein (RBP). Inside the cells, all-trans retinol and its oxidation product, all-trans retinal, are associated with different isoforms of cellular retinol-binding proteins (CRBP), while all-trans RA intracellularly binds to cellular retinoic acid-binding protein isoforms (CRABP).

2.1.4.3 RBP

Retinol is secreted from its storage pools and circulates in blood by binding to RBP. The main storage site for vitamin A and the main site of synthesis of RBP is the liver, although other tissues (including adipose tissue, kidney, lung, heart, skeletal muscle, spleen, eye and testis) also express this protein. Secretion of RBP from the liver is regulated by the availability of retinol [9]. Vitamin A deficiency inhibits RBP secretion, leading to protein accumulation in the endoplasmic reticulum of hepatic parenchymal cells. In the presence of retinol, RBP associates with retinol, moves to the Golgi apparatus and is secreted into blood. The mechanism by which retinol initiates RBP secretion from cells is not known. In blood, RBP is bound to the small protein transthyretin, which in addition to associating with RBP functions as a carrier protein for thyroid hormones. Binding of RBP to transthyretin prevents the loss of this smaller protein by filtration in the renal glomeruli. The transthyretin-RBP-retinol complex transports retinol in the circulation and delivers it to target tissues [10].

Important insights into the biological role of RBP have been obtained by studies of mice and humans in which the RBP gene is disrupted. RBP-deficient mice display both reduced blood retinol levels and impaired visual function during the first months of life. When maintained on a vitamin A-sufficient diet, they acquire normal vision by 5 months of age, even though their blood retinol level remains low. A striking phenotype of the RBP-null mice is that they possess larger than normal hepatic vitamin A storage, but are dependent on a continuous dietary intake of vitamin A [11], further proving the importance of RBP as a transporting protein. A study of two human siblings that harbored point mutations in their RBP gene and exhibited undetectable plasma RBP levels revealed that these sisters suffered from night blindness and mild retinal dystrophy but did not exhibit other clinical symptoms of vitamin A deficiency [12]. Taken together, RBP is critical for the mobilization of retinol from hepatic storage pools; however, RBP is not essential for the delivery of retinol to target tissues. Supply of vitamin A to target tissues in the absence of RBP is likely to be accomplished via newly absorbed retinyl esters or β-carotene present in circulating chylomicrons. Increased RBP has been shown to contribute to insulin resistance and type 2 diabetes [11]. All-trans RA has recently been shown to increase insulin sensitivity in diabetic mice while lowering RBP [13]. The effect on binding proteins must be considered when retinoids are used for disease treatment.

2.1.4.4 SRA6

The stimulated by retinoic acid gene 6 (STRA6) encodes the cell surface RBP receptor, which binds specifically to RBP and mediates retinol uptake from holo-RBP [14]. STRA6 is a widely expressed transmembrane protein. In mouse mammary epithelial cells, STRA6 expression can be up regulated by Wnt1 and retinoids. In addition, STRA6 mRNA levels are up regulated in mouse mammary gland tumors and human colorectal tumors [15]. Importantly, while the RBP-null mice and humans give rise to relative mild phenotypes, STRA6-null mice develop anophthalmia, congenital heart defects, diaphragmatic hernias, alveolar capillary dysplasia, lung hypoplasia, and mental retardation. These findings suggest that STRA6 may have additional functions that are not related to RBP transport [16].

2.1.4.5 CRBP

CRBPs belong to the family of fatty acid binding proteins in which expression of CRBP family members are tissue specific. For example, CRBP-II is expressed only in the enterocytes of the intestine, while CRBP-I and -III are expressed throughout embryonic and adult tissues [17]. Knockout studies for CRBP isoforms have identified differences in function due to altered tissue localization. CRBP-I knockout mice are healthy. However, they have low levels of hepatic retinyl esters [18], and their hepatic lipid droplets appear to be smaller and less abundant than in wild type littermates. CRBP-II-null mice have impaired retinol uptake, but they develop and reproduce normally under vitamin A-enriched diet, albeit with reduced retinol storage [19]. Reduction of vitamin A in the maternal diet of CRBP-II-null mice during gestation results in neonatal mortality immediately following birth [19]. CRBP-III null mice have impaired vitamin A incorporation into milk, but they are otherwise healthy [20]. CRBP-I and CRBP-III compensate for each other to maintain normal retinoid homeostasis, but the compensation is incomplete during lactation [20]. The binding affinity of CRBP-I towards retinol is about 100-fold higher than that of CRBP-II. They display a similar binding affinity towards retinal and CRBP-II associates with retinol and retinal with similar affinities. CRBPs, and especially CRBP-I with its high affinity for retinol, may sequester retinol from its ability to disrupt cell membranes. Epigenetic silencing of CRBP is a common event in human cancers [21]. Silencing CRBP reduces the availability of retinyl esters in the bloodstream and decreases the body’s ability to metabolize retinol [22].

2.1.4.6 CRABP

CRABP-I and -II have been identified with a high affinity for all-trans RA. In humans, these isoforms display 74 percent sequence identity and are highly conserved among species; however, these CRABP isoforms display different patterns of expression across cells and developmental stages. In adults, CRABP-I is expressed ubiquitously, while CRABP-II is only expressed in the skin, uterus, ovary, and the choroid plexus. Both CRABPs are widely expressed in the embryo, although they do not usually co-exist in the same cells. The biological functions of CRABPs are not completely understood. In mouse knockout stu dies, disruption of either CRABP-I or -II only display mild defects in limb development [23], which suggests CRABPs may be involved in generation of appropriate RA concentration gradients in the developing limb bud. Both CRABP isoforms are present in cytosol and nucleus and thus may deliver the ligand directly to the nuclear receptor. The differential role of these two binding proteins remains to be studied (reviewed in [24] and [25]). Increased CRABP-I expression may also contribute to RA resistance of cancer cells [26]. The effect of CRABP on cancer therapy deserves more attention.

2.1.4.7 Retinoic Acid Receptors

The major breakthrough in understanding RA’s function occurred upon identifying and cloning the receptors for RA [2728]. RA regulates gene expression by binding to its nuclear receptors, which in turn activates transcription of their downstream target genes. Thus, retinoids exert their biological functions primarily by regulating gene expression. This was predicted by Sporn and Roberts in 1983, when they wrote: “Ultimately, it would appear that the problem of the molecular mechanism of action of retinoids in control of differentiation and carcinogenesis is converging on one of the central problems of all biology, the control of gene expression.” [29]

2.1.4.8 RAR and RXR

Two distinct classes of receptors for retinoids have been identified: retinoic acid receptors (RAR) and retinoid × receptors (RXR). Each class of receptor contains three subtypes – α, β, and γ. RARs can be activated by both all-trans and 9-cis-RA, while, RXRs are exclusively activated by 9-cis RA. However, due to the conversion of all-trans to 9-cis RA, high concentrations (10−5 M) of all-trans RA can also activate gene transcription in cells transfected with RXRs [30].

RXRs can form homo- and heterodimers with other receptors. In fact, RXRs are promiscuous receptors forming heterodimers with many different kinds of receptors, which include receptors for fatty acids [peroxisomal proliferator activated receptors (PPAR)], bile acids [farnesoid × receptor (FXR)], oxysterols [liver × receptor (LXR)], xenobiotics [pregnane × receptor (PXR) and constitutive androstane receptor (CAR)], vitamin D [vitamin D receptor (VDR)], and RA (RAR). RXRs can also form homodimers. Hypervitaminosis A leads to bone fracture suggesting that vitamin A and D compete for the same receptor [31]. Within these heterodimers, RXRs can exist as both active and silent partners. When it serves as an active partner, 9-cis RA and the ligand for the heterodimeric partner can activate the heterodimer, and addition both ligands give synergistic induction in gene transcription. For example, RXR is an active partner for PPAR. Similarly, heterodimeric complexes of RXR with LXR or FXR also retain 9-cis RA responsiveness. Thus, RAs can regulate PPAR- and FXR-mediated pathways [32]. Recently, we demonstrated that RAs could also activate PXR-, VDR, and CAR-mediated signaling and thus regulated xenobiotic metabolism and potentially its own oxidation [3335]. When RXR serves as a silent partner, the heterodimer of RXR and its partner does not respond to RA. Regardless of their active or silent role, RXRs must be present in order to exert biological actions of various nuclear receptors. Using hepatocyte RXRα-deficient mice [3637], we have demonstrated that RXRα does play vital roles in xenobiotic (alcohol, acetaminophen) and endobiotic (fatty acid, cholesterol, amino acid, and carbohydrate) metabolism [3340]. Thus, RXR functions as an auxiliary factor and determines the effects of other hormones, making RXR a master regulator. The structure of nuclear receptors is summarized in recent review articles [738].

Existing data suggest that the binding protein and receptor work together to exert the specific effect of RAs. For example, RAs can bind to both PPARβ, the receptor for fatty acids, and RAR. Fatty acid-binding protein 5 (FABP5) and CRABP-II are specific binding proteins that channel RAs from the cytosol into the nucleus for binding to either PPARβ or RAR, respectively [39]. The ratio of FABP5/CRABP-II concentrations determines which receptor is activated. By activating PPARβ, RAs induce expression of genes affecting lipid and glucose homeostasis, such as the insulin-signaling gene pyruvate dehydrogenase kinase 1 (PDK1), which enhances insulin action. Hence, RAs stimulate lipolysis and reduce triglyceride content. RA implantation into obese mice causes up regulation of PPARβ as well as an increased expression of PPARβ target genes, including PDK1, which led to weight loss [40].

2.1.4.9 Retinoids and Cancer

Retinoids are widely used to treat visual and dermatological diseases. Their effect on cancer prevention and treatment has received a lot of attention. This review focuses on the action of retinoids on cancer. Retinoids have been used as potential chemotherapeutic or chemopreventive agents because of their differentiation, anti-proliferative, pro-apoptotic, and anti-oxidant effects. Epidemiological studies show that lower vitamin A intake results in a higher risk of developing cancer, which aligns with observations of vitamin A-deficient animals [61]. Altered expression of RA receptors is also associated with malignant transformation of animal tissues or cultured cells. Furthermore, retinoids suppress carcinogenesis in tumorigenic animal models for skin, oral, lung, breast, bladder, ovarian, and prostate [6268]. In humans, retinoids reverse premalignant human epithelial lesions, induce the differentiation of myeloid cells, and prevent lung, liver, and breast cancer [6973].

The following is a summary of how major retinoids may work in cancer treatment or prevention.

2.1.4.9.1 All-trans RA (tretinoin)

All-trans RA is the most abundant natural retinoid and has been widely studied for many years. It is currently in clinical trials for the treatment of lymphoma, leukemia, melanoma, lung cancer, cervical cancer, kidney cancer, neuroblastoma, and glioblastoma. The most effective clinical usage of all-trans RA in human disease was demonstrated in treatment of a rare leukemia, acute promyelocytic leukemia (APL). APL is characterized by selected expansion of immature myeloid precursors or malignant myeloid cells blocked at the promyelocytic stage of hemopoietic development. APL cells invariably express aberrant fusion proteins involving the DNA and ligand binding domain of RARα [7475]. Other fusion partners include the promyelocytic leukemia zinc finger gene, the nucleophosmin gene, the nuclear mitotic apparatus gene, and the Stat5b gene, while the most common fusion partner is promyelocytic leukemia protein (PML). The PML-RARα chimeric receptor is created by a balanced reciprocal chromosomal translocation, t(15;17)(q22:q11). The expressed PML-RARα chimeric receptor alters normal function of RARs. PML-RARα can form a homodimer through the coiled-coil motif of PML, inhibiting RARα’s ability to bind to RA responsive elements, thereby preventing activation of downstream target genes [7677]. In addition, RXR is an essential component of the oncogenic PML/RARα complex suggesting RXR can be a drug target for APL [7879]. In 1995, the FDA approved all-trans RA for treating APL. The all-trans RA-induced differentiation of APL cells is due to both its ability to promote the degradation of the mutant PML-RARα and the dissociation of its co-repressors [80]. All-trans RA also causes cell cycle arrest at G1 phase and inhibits cell proliferation [81]. In addition, high concentration of all-trans RA induces post-maturation apoptosis of APL-blasts through the induction of the tumor-selective death ligand tumor necrosis factor-related apoptosis-inducing ligand TRAIL [82].

RA syndrome is a life-threatening complication seen in APL patients treated with all-trans RA. This syndrome is characterized by dyspnea, fever, weight gain, hypotension, and pulmonary infiltrates. It can be effectively treated by giving dexamethasone and holding off all-trans RA treatment in severe cases. An elevated white count is sometimes associated with this syndrome, but is not a prerequisite. The etiology of RA syndrome is not clear; several causes have been speculated including a capillary leak syndrome from cytokine release from the differentiating myeloid cells. Alternatively, all-trans RA may cause the maturing myeloid cells to acquire the ability to infiltrate organs such as the lung [83].

2.1.4.9.2. 9-cis RA (alitretinoin)

9-cis RA differentiates itself from all-trans RA in its ability to activate both RAR and RXR. In addition, 9-cis RA activates PPAR, FXR, PXR, VDR, and CAR via RXR. In preclinical studies, 9-cis RA is effective in the prevention of mammary and prostate cancer [8485] and it has also been FDA-approved for the topical treatment of cutaneous lesions of Kaposi’s sarcoma [86]. In addition, 9-cis RA and all-trans RA can individually induce apoptosis of human liver cancer cells [87]. 9-cis RA not only regulates nuclear genes, but also mitochondria gene transcription [88].

2.1.4.9.3. 13-cis RA (isotretinoin)

13-cis RA is unique that it exhibits immunomodulatory and anti-inflammatory responses. It inhibits ornithine decarboxylase, thereby decreasing polyamine synthesis and keratinization [89]. 13-cis RA noticeably reduces the production of sebum and shrinks the sebaceous glands [90]. It stabilizes keratinization and prevents comedones formation [9192]. The exact mechanism of action is unknown. This combination of regulating proliferation, differentiation, and inflammation could make 13-cis RA a more effective drug in comparison to other retinoids, which may cause inflammation and irritation [93].

13-cis RA is in clinical trial for different types of cancers, and thyroid cancer received a lot of attention. In follicular thyroid cancer cells, 13-cis RA induces radioiodine avidity of cells formerly unable to accumulate radioiodine [94]. In human thyroid carcinoma cell lines, retinoids induce the expression of type I iodothyronine-5′-deiodinase and sodium/iodide-symporter, which are the thyroid differentiation markers [95]. However, approximately 30% of thyroid tumors dedifferentiate after treatment and thus develop into highly malignant anaplastic thyroid carcinomas [96]. 13-cis RA is also used to treat non-operable thyroid follicular tumors, which fail to uptake radioiodine. 13-cis RA increases the radioiodide uptake in some patients. The beneficial outcome of this treatment was interpreted as partial re-differentiation of thyroid cancer cells. This effect of 13-cis RA requires the existence of functional RXR [96]. The effect of 13-cis RA on thyroid cancer has been reviewed extensively [97]. Besides thyroid cancer, utilizing 13-cis RA for maintenance therapy has significantly improved the outcome of patients with a high-risk form of neuroblastoma [98]. Along the same line of work, Krüppel zinc-finger protein ZNF423 is critical for RA signaling and is likely a prognostic marker for neuroblastoma [99]. 13-cis RA is also effective in preventing head and neck cancer, which is discussed below.

2.1.4.9.4. Synthetic Retinoids

N-(4-hydroxyphenyl) retinamide (Fenretinide or 4HPR) was first synthesized in the late 1960s by R. W. Pharmaceuticals. Since then, the biological properties of fenretinide have been of great interest. Currently, fenretinide is one of the most promising clinically tested retinoids. The modification of the carboxyl end of all-trans RA with an N-4-hydroxyphenyl group resulted in increased efficacy as a chemoprevention agent as well as reduced toxicity when compared with other retinoids [100]. Animal models have demonstrated that treatment with fenretinide prevents chemically induced cancers of the breast, prostate, bladder, and skin [101104]. Furthermore, the combination of tamoxifen with fenretinide produces efficacy greater than either chemical alone [105].

Natural retinoids like all-trans RA induce differentiation and/or cytostasis in target cells [106108], while fenretinide has distinct biologic effects including the induction of apoptosis by generating reactive oxygen species (ROS) and lipid second messengers [104]. The apoptotic effect of fenretinide has been documented in a variety of cancer cells including transformed T cells, B cells and breast epithelial cells, as well as bladder, breast, cervical, colon, embryonal, esophageal, head and neck, lung, ovarian, pancreatic, prostate, and skin carcinomas [100]. Furthermore, fenretinide does not induce point mutations or chromosomal aberrations, and is therefore not genotoxic [109]. These qualities suggest that fenretinide could be used for a long-term chemopreventive modality. In animal models, fenretinide has demonstrated chemopreventive efficacy against carcinogenesis of the breast [110], prostate, pancreas, and skin [104111112]. Moreover, in a clinical setting, fenretinide slowed the progression of prostate cancer in men diagnosed with an early stage of the disease [113]. Fenretinide protected against the development of ovarian cancer and a second breast malignancy in premenopausal women who had been treated to prevent the progression of early-stage breast cancer [114]. It also prevented relapse and the formation of secondary primary lesions in patients following the surgical removal of oral leukoplakia [115]. Recent studies also illustrated the anti-angiogenic [116] and anti-fibrotic [117] effect of fenretinide. Furthermore, long-term fenretinide treatment prevents high-fat diet-induced obesity, insulin resistance, and hepatic steatosis [118].

The mechanisms associated with fenretinide-induced apoptosis have been explored, but are not well-understood [100]. The components that lead to ROS generation and cause cell death are largely unknown. Depending on cell types and models used, the effect of fenretinide has been shown to be RARβ-dependent or -independent [119]. Our data showed that fenretinide-induced apoptosis of human liver cancer cells was RARβ-dependent [120]. Furthermore, induction and cytoplasmic localization of Nur77 dictates the sensitivity of liver cancer cell to fenretinide-induced apoptosis [121]. It seems that fenretinide enriches the cytoplasmic Nur77 to target mitochondria and induce cell death. The relationship between RARβ and Nur77 in mediating fenretinide-induced apoptosis remains to be determined.

A retinoid-related molecule 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalenecar-boxylic acid (AHPN) (also called CD437) and it’s analog (E)-4-[3-(1-adamantyl)-4-hydroxyphenyl]-3-chlorocinnamic acid (3-Cl-AHPC) also have Nur77-dependent apoptotic effects [122124]. AHPN is structurally distinct from fenretinide. AHPN-induced apoptosis activates JNK [125127], which is required for maximal apoptosis induction and precedes mitochondrial depolarization. Induction of apoptosis of breast and prostate cancer cells by AHPN is also associated with its inhibition of Akt activity [128]. Thus, induction of JNK and inhibition of Akt phosphorylation of Nur77 contribute to Nur77 nuclear export mediated by AHPN [129].

While many synthesized RAs are promising for cancer treatment, only a few are FDA-approved or currently undergoing clinical trials for cancer therapy. A number of retinoids, which have been FDA-approved for dermatological purposes, have potential for cancer treatment. Bexarotene (Targretin) is a synthetic retinoid approved by the FDA to treat skin problems caused by cutaneous T-cell lymphoma that are unresponsive to other treatments [130]. Other synthetic retinoids, such as TAC-101 (Taiho Pharmaceutical, Tokyo, Japan) has shown efficacy in inhibiting tumor growth in the liver and markedly increases survival in both the primary HCC and metastatic colon cancer models [131]. TAC-101 is currently in phase II trial for hepatocellular carcinoma and has shown good preliminary success [132]. Another, Tazarotene (AVAGE) (Allergan, Irvine, CA) is in phase I trials for the treatment of lymphoma [133]. Please see table 1 for a brief characterization of some of retinoids that are in use or in clinical trials.

3  Vitamin C

3.1 American Cancer Society

http://www.cancer.org/treatment/treatmentsandsideeffects/complementaryandalternativemedicine/herbsvitaminsandminerals/vitamin-c

Vitamin C is an essential vitamin the human body needs to function well. It is a water-soluble vitamin that cannot be made by the body, and must be obtained from foods or other sources. Vitamin C is found in abundance in citrus fruits such as oranges, grapefruit, and lemons, and in green leafy vegetables, tomatoes, potatoes, strawberries, red or green peppers, and cantaloupe.

Vitamin C is found in many vegetables and fruits, especially oranges, grapefruits, and peppers. Many studies have shown a link between eating foods rich in vitamin C, such as fruits and vegetables, and a reduced risk of cancer. On the other hand, the few studies in which vitamin C has been given as a supplement have not shown a reduced cancer risk.

This suggests that the activity of fruits and vegetables in preventing cancer is due to a combination of many things such as vitamins, fiber, and other phytochemicals and not to vitamin C alone (see Phytochemicals). Clinical trials of high doses vitamin C as a treatment for cancer have not shown any benefit. High doses of vitamin C can cause side effects in some people.

3.2 Intravenous vitamin C for cancer

Oct 4, 2013 | By Dr. Ronald Hoffman

For more than 20 years, the Hoffman Center has been using high-dose vitamin C drips in its cancer support protocols. The initial impetus was from Linus Pauling who, together with Ewan Cameron, pioneered the use of high-dose C in cancer in the 1960s.

Now, there’s new interest in this modality for fighting cancer based on new, exciting research under way at the National Institutes of Health.

Cameron and Pauling found that vitamin C helped cancer patients live about four times longer than cancer patients not given vitamin C. They administered high-dose vitamin C in the form of sodium ascorbate given orally and intravenously to treat more than 1,000 cancer patients.

Nonetheless, vitamin C for cancer suffered a setback when Dr. Charles Moertel of the Mayo Clinic, an arch foe of nutritional therapies for cancer, sought to disprove Pauling’s thesis. But he did not follow the Pauling/Cameron instructions or regimen.

Moertel selected a cohort of terminal colon cancer patients who had not responded to all forms of conventional treatment, including surgery, chemo and radiation, and administered 10 grams of vitamin C to them orally. When the patients failed to demonstrate improved survival over patients not receiving vitamin C in the study, Moertel pronounced the vitamin C/cancer hypothesis defunct.

Moertel failed to note that the benefits achieved by Pauling and Cameron’s patients were obtained via both IV and oral C. He ultimately succumbed to cancer himself years later.

Alternative practitioners, meanwhile, sought to resurrect IV vitamin C as a tool in the treatment of cancer, but not until recently has serious academic research resumed.

Dr. Hugh Riordan of Kansas treated hundreds of cancer patients with doses of vitamin C up to 200,000 mg (200 grams) per day in infusions lasting 4-12 hours several times a week. He compiled a series of case histories documenting impressive responses but passed recently, before his work was generally acknowledged.

His protegee, Dr. Jeanne Drisko, Director, KU Integrative Medicine, has undertaken a series of clinical trials to validate the benefits of IV vitamin C in cancer. An FDA approved trial is now underway.

Research at the National Institutes of Health is beginning to suggest that vitamin C deserves another chance to find its niche in the arsenal of anti-cancer therapies. Studies now suggest that even high dose vitamin C given by mouth is poorly absorbed. Blood levels “max out” at doses of 500 mg given several times during the day.

But vitamin C given intravenously is another story. When delivered in a “drip,” much higher concentrations of C can be attained. At these higher concentrations, vitamin C has different characteristics than if given orally. While oral vitamin C boosts immunity and assists tissue repair, it is too weak to do much to kill or inhibit cancer cells. But at high doses delivered directly into the bloodstream, it may act to increase levels of hydrogen peroxide deep in the tissues where cancer cells lurk. Peroxide-mediated killing is one of the white blood cells’ key mechanisms for fighting infection and cancer.

Research currently under way has shown that high concentrations of vitamin C can stop the growth or even kill a wide range of cancer cells. Only intravenous administration of vitamin C can deliver the high doses found to be effective against cancer.

IV vitamin C, when administered by a trained, experienced physician, is safe and well-tolerated, even at doses as high as 100,000 mg (100 grams) per day. Proper blood tests must be done to ensure that it is well-tolerated, and the patient must be monitored. Doses must be gradually adjusted upward. Not all patients are candidates for IV vitamin C. Vitamin C can be safely administered even while patients are undergoing chemo and radiation; in fact, the FDA-approved trial at the University of Kansas Medical Center explicitly permits the co-administration of vitamin C with conventional treatments.

3.3 IV Vitamin C Kills Cancer Cells

by Dr. Julian Whitaker

By now, most people know that vitamin C is a potent antioxidant that has the power to boost immune function, increase resistance to infection, and protect against a wide range of diseases.

But there’s an entirely different and largely unknown role of vitamin C, and that is its ability—when administered in very high doses by intravenous (IV) infusions—to kill cancer cells.

Vitamin C interacts with iron and other metals to create hydrogen peroxide. In high concentrations, hydrogen peroxide damages the DNA and mitochondria of cancer cells, shuts down their energy supply, and kills them outright. Best of all—and unlike virtually all conventional chemotherapy drugs that destroy cancer cells—it is selectively toxic. No matter how high the concentration, vitamin C does not harm healthy cells.

Lab studies reveal that this therapy is effective against many types of cancer, including lung, brain, colon, breast, pancreatic, and ovarian. Animal studies show that when human cancers are grafted into animals, high-dose IV vitamin C decreases tumor size by 41 to 53 percent “in diverse cancer types known for both their aggressive growth and limited treatment options.” Additionally, numerous patient case reports have been written up in medical journals.

Why IV Administration Is Essential

The only way to get blood levels of vitamin C to the concentrations required to kill cancer cells is to administer it intravenously. The body tightly controls levels of this vitamin by limiting intestinal absorption. If you took 10 g (10,000 mg) of vitamin C by mouth at one time, you would only absorb around 500 mg—and you’d get a serious case of diarrhea!

Intravenous administration, however, bypasses this control mechanism, and blood levels rise in a dose-dependent manner. For example, 10 g of IV vitamin C raises blood levels 25 times higher than the same dose taken orally, and this increases up to 70-fold as doses get larger.

4 Expert Q&A: Vitamin D and Cancer Risk

http://www.cancer.net/navigating-cancer-care/prevention-and-healthy-living/diet-and-nutrition/expert-qa-vitamin-d-and-cancer-risk

Vitamin D is one of several nutrients that the body needs to stay healthy. It may also play a role in reducing the risk of cancer, and several research studies are exploring this link. Cancer.Net talked with Richard Goldberg, MD, to learn more about current research on vitamin D and what people should know.

Q: What is the role of vitamin D in the body, and what are some sources of this vitamin?

A: One role of vitamin D is to regulate the absorption of calcium by the body. Calcium is the main component of bones and is important in the function of all cells in the body, particularly the heart. People who are vitamin D deficient (don’t get enough) can have weakened bones (a condition called osteoporosis in adults and rickets or osteomalacia in children). Too little calcium (called hypocalcemia) in the body can lead to irregular heartbeat and muscle spasms.

Milk, fish, eggs, and fortified cereals and orange juice are good sources of vitamin D. Milk manufactured in the United States is generally fortified with vitamin D as a way to prevent deficiencies from occurring. Supplemental vitamins are also a source.

Unlike other vitamins that the body cannot produce by itself, vitamin D can either be absorbed directly from the intestine or made from compounds in foods. The body can make vitamin D from nutrients related to cholesterol. These nutrients are then converted to vitamin D as they circulate in the blood when a person’s skin is exposed to sunlight.

Too much vitamin D can also be bad for a person, leading to drowsiness, kidney stones, bone or muscle weakness, and elevated blood calcium, a condition called hypercalcemia that can cause confusion and, in extreme cases, death.

Q: When getting vitamin D from sunlight, how long should a person be exposed to the sun? What are the risks of too much sun exposure?

A: While 90% of the body’s vitamin D comes from exposure to sun (in the absence of vitamin D supplements), the amount of sun exposure needed to produce adequate vitamin D levels is actually quite limited. Sun exposure at the equator is far more intense than in such northern cities as Boston or London, for instance, and is more intense anywhere in summer than in winter. However, it takes only five to ten minutes of exposing the hands and face three times a week to receive adequate sun exposure in the summer in Boston. Exposure of more skin, such as when wearing a bathing suit, requires only a very short time in the sun. Use of sunblock is very important when sun exposure is longer than that to prevent skin cancer, including melanoma, and other sun-induced damage such as wrinkling and pigmentation changes (sunspots). Learn more about protecting your skin from the sun.

Q: How might vitamin D work to help lower the risk of cancer?

A: Laboratory studies have shown that vitamin D deficiency can lead to decreased communication between cells and leads them to stop sticking to one another, a condition that could cause cancer cells to spread. Compared with normal cells, cancer cells remain in an immature state, and vitamin D appears to have a role in making cells mature. Vitamin D also appears to play a role in regulating cellular reproduction, which malfunctions (doesn’t work properly) in cancer. Higher levels of vitamin D lead to cellular adherence, maturation, and communication between cells, all of which may lower cancer risk.

Q: What does research show about vitamin D levels and cancer?

A: Studies in populations have shown that low vitamin D levels are a risk factor for cancer in general, and particularly for prostatecolorectal, and breast cancers.

There are also data that correlate high blood levels of vitamin D with a reduced risk of breast and colorectal cancers. These levels can best be achieved by taking supplemental vitamin D. In colorectal cancer, calcium supplementation may also reduce the risk of polyps (noncancerous growths that may develop on the inner wall of the colon and rectum) and cancer. Numerous studies have tested cancer risk by giving patients supplemental vitamin D, with or without calcium supplementation. While the results are somewhat variable, substantial reduction (on the order of 50%) in the odds of breast and colon cancers with supplementation, have been noted in some studies. People with a personal history of these types of cancer and their relatives may wish to discuss supplementation with their doctors.

5 Magnesium and Cancer Research

5.1  Dr Sircus on Mar 18, 2010
http://drsircus.com/medicine/magnesium/magnesium-and-cancer

Aleksandrowicz et al in Poland conclude that inadequacy of magnesium and antioxidants are important risk factors in predisposing to leukemias.[2] Other researchers found that 46% of the patients admitted to an ICU in a tertiary cancer center presented hypomagnesemia. They concluded that the incidence of hypomagnesemia in critically ill cancer patients is high.[3]In animal studies we find that magnesium deficiency has caused lymphopoietic neoplasms in young rats. A study of rats surviving magnesium deficiency sufficient to cause death in convulsions during early infancy in some, and cardiorenal lesions weeks later in others, disclosed that some of survivors had thymic nodules or lymphosarcoma.[4]

One would not normally think that Magnesium (Mg) deficiency can paradoxically increase the risk of, or protect against cancer yet we will find that just as severe dehydration or asphyxiation can cause death magnesium deficiency can directly lead to cancer. When you consider that over 300 enzymes and ion transport require magnesium and that its role in fatty acid and phospholipids acid metabolism affects permeability and stability of membranes, we can see that magnesium deficiency would lead to physiological decline in cells setting the stage for cancer. Anything that weakens cell physiology will lead to the infections that surround and penetrate tumor tissues. These infections are proving to be an integral part of cancer. Magnesium deficiency poses a direct threat to the health of our cells. Without sufficient amounts our cells calcify and rot. Breeding grounds for yeast and fungi colonies they become, invaders all too ready to strangle our life force and kill us.

Over 300 different enzymes systems rely upon magnesium to facilitate their catalytic action, including ATP metabolism, creatine-kinase activation, adenylate-cyclase, and sodium-potassium-ATPase.[5]

It is known that carcinogenesis induces magnesium distribution disturbances, which cause magnesium mobilization through blood cells and magnesium depletion in non-neoplastic tissues. Magnesium deficiency seems to be carcinogenic, and in case of solid tumors, a high level of supplemented magnesium inhibits carcinogenesis.[6] Both carcinogenesis and magnesium deficiency increase the plasma membrane permeability and fluidity. Scientists have in fact found out that there is much less Mg++ binding to membrane phospholipids of cancer cells, than to normal cell membranes.[7]

Magnesium protects cells from aluminum, mercury, lead, cadmium, beryllium and nickel.

Magnesium in general is essential for the survival of our cells but takes on further importance in the age of toxicity where our bodies are being bombarded on a daily basis with heavy metals.Glutathione requires magnesium for its synthesis.[8] Glutathione synthetase requires ?-glutamyl cysteine, glycine, ATP, and magnesium ions to form glutathione.[9] In magnesium deficiency, the enzyme y-glutamyl transpeptidase is lowered.[10] According to Dr. Russell Blaylock, low magnesium is associated with dramatic increases in free radical generation as well as glutathione depletion and this is vital since glutathione is one of the few antioxidant molecules known to neutralize mercury.[11]Without the cleaning and chelating work of glutathione (magnesium) cells begin to decay as cellular filth and heavy metals accumulates; excellent environments to attract deadly infection/cancer.

There is drastic change in ionic flux from the outer and inner cell membranes both in the impaired membranes of cancer, and in Mg deficiency.

Anghileri et al[12],[13] proposed that modifications of cell membranes are principal triggering factors in cell transformation leading to cancer. Using cells from induced cancers, they found that there is much less magnesium binding to membrane phospholipids of cancer cells, than to normal cell membranes.[14] It has been suggested that Mg deficiency may trigger carcinogenesis by increasing membrane permeability.[15] Magnesium deficient cells membranes seem to have a smoother surface than normal, and decreased membrane viscosity, analogous to changes in human leukemia cells.[16],[17] There is drastic change in ionic flux from the outer and inner cell membranes (higher Ca and Na; lower Mg and K levels), both in the impaired membranes of cancer, and of Mg deficiency. And we find that lead (Pb) salts, are more leukemogenic when given to Mg deficient rats, than when they are given to Mg-adequate rats, suggesting that Mg is protective.[18]

Magnesium has an effect on a variety of cell membranes through a process involving calcium channels and ion transport mechanisms. Magnesium is responsible for the maintenance of the trans-membrane gradients of sodium and potassium.

Long ago researchers postulated that magnesium supplementation of those who are Mg deficient, like chronic alcoholics, might decrease emergence of malignancies[19] and now modern researchers have found that all types of alcohol — wine, beer or liquor — add equally to the risk of developing breast cancer in women. The researchers, led by Dr. Arthur Klatsky of the Kaiser Permanente Medical Care Program in Oakland, Calif., revealed their findings at a meeting of the European Cancer Organization in Barcelona in late 2007. It was found that women who had one or two drinks a day increased their risk of developing breast cancer by 10 percent. Women who had more than three drinks a day raised their risk by 30 percent. The more one drinks the more one drives down magnesium levels.

Breast cancer is the second most common cancer killer of women, after lung cancer. It will be diagnosed in 1.2 million people globally this year and will kill 500,000.

According to data published in the British Journal of Cancer in 2002, 4 percent of all breast cancers — about 44,000 cases a year — in the United Kingdom are due to alcohol consumption. It’s an important question though, and one not asked by medical or health officials, is it the alcohol itself or the resultant drop in magnesium levels that is cancer provoking? Though some studies have shown that light- to moderate alcohol use can protect against heart attacks it does us no good to drink if it causes cancer. Perhaps if magnesium was supplemented in women drinkers who were studied there would have been no increase of cancer from drinking.

Alcohol has always been known to deplete magnesium, and is one of the first supplements given to alcoholics when they stop and attempt to detoxify and withdraw.

Researchers from the School of Public Health at the University of Minnesota have just concluded thatdiets rich in magnesium reduced the occurrence of colon cancer.[20] A previous study from Sweden[21] reported that women with the highest magnesium intake had a 40 per cent lower risk of developing the cancer than those with the lowest intake of the mineral.

Magnesium stabilizes ATP[22], allowing DNA and RNA transcriptions and repairs.[23]

The anti-colon cancer effects of calcium are linked to magnesium levels, says a new study. Researchers from Vanderbilt University found that low ratios of the minerals were associated with reduced risk of colorectal cancer, according to findings presented at the Seventh Annual American Association for Cancer Research International Conference on Frontiers in Cancer Prevention Research. Both high magnesium and calcium levels have been linked to reduced risks of colon cancer but studies have also shown that high calcium levels inhibit the absorption of magnesium. According to Qi Dai, MD, PhD, and co-workers, Americans have high calcium intake, but also a high incidence of colorectal cancer. “If calcium levels were involved alone, you’d expect the opposite direction. There may be something about these two factors combined – the ratio of one to the other – that might be at play,” said Dai. The risk of colorectal cancer adenoma recurrence was reduced by 32 per cent among those with baseline calcium to magnesium ratio below the median in comparison to no reduction for those above the median,” said Dai.[24]

Pre-treatment hypomagnesemia has been reported in young leukemic children, 78% of whom have histories of anorexia, and have excessive gut and urinary losses of Mg.[25]

Several studies have shown an increased cancer rate in regions with low magnesium levels in soil and drinking water, and the same for selenium. In Egypt the cancer rate was only about 10% of that in Europe and America. In the rural fellah it was practically non-existent. The main difference was an extremely high magnesium intake of 2.5 to 3g in these cancer-free populations, ten times more than in most western countries.[26]

5.2 Magnesium and cancer: a dangerous liason.

Castiglioni S, Maier JA.
Magnes Res. 2011 Sep; 24(3):S92-100
http://dx.doi.org:/10.1684/mrh.2011.0285

A complex relationship links magnesium and cancer. The aim of this review is to revisit current knowledge concerning the contribution of magnesium to tumorigenesis, from transformed cells to animal models, and ending with data from human studies. Cultured neoplastic cells tend to accumulate magnesium. High intracellular levels of the cation seem to confer a metabolic advantage to the cells, contribute to alterations of the genome, and promote the acquisition of an immortal phenotype. In magnesium-deficient mice, low magnesium both limits and fosters tumorigenesis, since inhibition of tumor growth at its primary site is observed in the face of increased metastatic colonization. Epidemiological studies identify magnesium deficiency as a risk factor for some types of human cancers. In addition, impaired magnesium homeostasis is reported in cancer patients, and frequently complicates therapy with some anti-cancer drugs. More studies should be undertaken in order to disclose whether a simple and inexpensive intervention to optimize magnesium intake might be helpful in the prevention and treatment of cancer.

Even though cancer-associated death rates are falling steadily, the global burden of cancer continues to increase primarily as a result of an aging population, but also because of the adoption of cancer-causing behaviors, including smoking and a western-type diet [1]. In particular, statistical and epidemiological data point to diet as responsible for about 35% of human cancer mortality [2]. There is general agreement about the inverse correlation between the risk of cancer and the regular consumption of fruit, cereals and vegetables, rich sources of many beneficial micronutrients, vitamins and minerals. Magnesium, which is predominantly obtained by eating unprocessed grains and green leafy vegetables, is an essential micronutrient implicated in a wide variety of regulatory, metabolic and structural activities [3]. The occidental diet is relatively deficient in magnesium Presented in part at the European Magnesium Meeting – EUROMAG Bologna 2011, San Giovanni in Monte, Bologna, Italy, June 8-10, 2011. because of the processing of many food items and the preference for calorie-rich, micronutrient-poor foods [4]. Magnesium deficiency complicates chronic gastrointestinal and renal diseases, diabetes mellitus, alcoholism, and therapies with some classes of diuretics and anticancer drugs [4]. A review of the literature reveals the relationship between magnesium and cancer, from the cellular level through to animal models and humans. Although controversy exists about the role of magnesium in tumors, most of the results available point to low magnesium as a factor contributing to tumorigenesis.

5.1.1 Magnesium acts as a secondary messenger, and activates a vast array of enzymes [3, 5]. Since magnesium participates in all major metabolic processes, as well as redox reactions, it is no surprise that it has a direct role in controlling cell survival and growth. In normal diploid cells, the total concentration of magnesium increases throughout the G1 and S phases of the cell cycle. Accordingly, low extracellular magnesium markedly inhibits their proliferation [3]. Conversely, neoplastic cells are refractory to the proliferative inhibition by low extracellular magnesium but, being extremely avid for the cation, it accumulates in these cells even when cultured in low magnesium levels [6]. This avidity is due, at least in part, to an impairment of Na-dependent magnesium extrusion [7], and to the overexpression of one of the magnesium transporters, namely transient receptor potential melastatin (TRPM)7 [8]. High intracellular magnesium seems to provide a selective advantage for the transformed cells since magnesium contributes to regulating enzymes of various metabolic pathways and of the systems involved in DNA repair. Indeed, magnesium forms complexes with ATP, ADP and GTP, necessary for the activity of enzymes implicated in the transfer of phosphate groups such as glucokinase, phosphofructokinase, phosphoglycerate kinase and pyruvate kinase [9], enzymes of glycolysis known to be the pathway used preferentially by neoplastic cells to produce energy [10]. Magnesium also forms complexes with DNA polymerase, ribonucleases, adenylcyclase, phosphodiesterases,guanylate-cyclase, ATPases and GTPases, being therefore implicated in the metabolism of nucleic acids and proteins, and in signal transduction [9]. Since mutation is a driving force in the development of cancer, it is worth noting that magnesium is involved in the inhibition of N-methylpurine DNA-glycosidase, which initiates base excision repair in DNA by removing a wide variety of alkylated, deaminated, and lipid peroxidation-induced purine adducts [11]. In addition, the nuclear Ser/Thr phosphatase PPM1D (also known as WIP1), which is overexpressed in various human primary tumors, requires magnesium for its activity. PPM1D is involved in the regulation of several essential signaling pathways implicated in tumorigenesis [12, 13]. In particular, PPM1D dephosphorylates and, therefore, inactivates the p53 tumor suppressor gene, a canonical suppressor of proliferation. It also complements several oncogenes, such as Ras, Myc, and HER-2/neu, for cellular transformation both in vitro and in vivo [12].

On these bases, it is possible to conclude that high intracellular magnesium has a role in promoting genetic instability. Another peculiarity of tumor cells is their limitless proliferative potential [14, 15]. It is therefore relevant to point out that magnesium is required to activate telomerase [16-18], a specialized DNA polymerase that extends telomeric DNA and counters the progressive telomere erosion associated with cell duplication. The presence of telomerase activity correlates with a resistance to induction of both senescence and apoptosis which are considered to be crucial anticancer defenses [14, 15]. These points are summarized in figure 1, which also underlines the contribution of high intracellular magnesium to some of the hallmarks of cancer, as highlighted by Hanahan and Weinberg [14, 15]. Mentioning only studies performed on neoplastic cells would be simplistic, since tumors are more than just masses of proliferating cancer cells. Rather, they are complex, heterotypic tissues where normal cells in the stroma, far from being passive bystanders, actively collaborate to cancer development and progression [14, 15]. Many of the growth signals driving the proliferation of and invasion by carcinoma cells originate from the stromal cell components of the tumor mass. It is therefore worth noting that low magnesium modulates the functions of a variety of normal cells present in the tumor microenvironment. In particular, endothelial cells cultured in low magnesium release higher amounts of metalloproteases and growth factors [19]. Similar results were obtained in cultured human fibroblasts (unpublished results). In addition, low magnesium promotes endothelial and fibroblast senescence [20], and senescent cells can modify the tissue environment in a way that synergizes with oncogenic mutations to promote the progression of cancers [21]. Only the behavior of microvascular endothelial cells cultured in low magnesium seems not to fit with the picture described above. It is well known that angiogenesis is crucial to nourish the tumor and facilitate its spreading, but low extracellular magnesium impairs acquisition of the angiogenic phenotype by microvascular endothelial cells. Exposure to low magnesium retards endothelial proliferation, migration and differentiation in vitro ([22] and manuscript submitted). Accordingly, magnesium-deficient mice develop tumors which are significantly less vascularized than the controls [23].

Figure 1. Neoplastic cells tend to have high intracellular concentrations of magnesium, which contribute to the regulation of various metabolic pathways and of systems involved in DNA repair, thus providing a selective advantage for the transformed cells. The figure also links the effects of high intracellular concentrations of magnesium on cell functions to some hallmarks of cancer as highlighted by Hanahan and Weinberg [14, 15].

5.1.2 Low magnesium and cancer: a focus on human studies

Several epidemiological studies have provided evidence that a correlation exists between dietary magnesium and various types of cancer. High levels of magnesium in drinking water protect against oesophageal and liver cancer [36, 37]. In addition, magnesium concentration in drinking water is inversely correlated with death from breast, prostate, and ovarian cancers, whereas no correlation existed for other tumors [36, 38, 39]. Epidemiological studies conducted in various countries demonstrate an association between low intake of magnesium and the risk of colon cancer [40-43]. In addition, a large population-based prospective study in Japan shows a significant inverse correlation between dietary intake of magnesium and colon cancer in men but not in women [44]. Intriguingly, the association between low intake of magnesium and colon cancer is linked to the increased formation of N-nitroso compounds, most of which are potent carcinogens [43]. A further link between magnesium and colon neoplasia is highlighted by the association of adenomatous and hyperplastic polyps, which might progress to carcinoma, with a genetic polymorphism of TRPM7 [45], an ubiquitous ion channel with a central role in magnesium uptake and homeostasis [46]. Results concerning the contribution of magnesium to lung cancer are controversial. A first case-control study correlates low dietary magnesium with increased lung cancer risk both in men and women [47]. This link is more evident in the elderly, current smokers, drinkers and in those with a late-stage disease. To explain the protective effect of magnesium against lung cancer, the authors recall that magnesium regulates cell multiplication, protects against the oxidative stress invariably associated with magnesium deficiency [48], and maintains genomic stability. A recent prospective analysis however, does not support the previous report [49]. These contrasting data could result from recall bias, the difficulty in evaluating diet composition and the fact that smoking is a very strong risk factor for lung cancer.

Conclusion Although the evidence is still fragmentary, most of the data available point to magnesium as a chemopreventive agent, so that optimizing magnesium intake might represent an effective and low-cost preventive measure to reduce cancer risk. Doubts remain about supplementing cancer patients with magnesium. The recently revived interest in the relationship between magnesium and tumors, both in experimental and clinical oncology, should encourage more studies that would advance our understanding of the role of magnesium in tumors, and could explore the possibility that optimizing magnesium homeostasis might prevent cancer or help in its treatment.

5.3 A Magnesium Deficiency Increases Cancer Risk Significantly

Wed, May 21, 2008 by: Mark Sircus

http://www.naturalnews.com/023279_magnesium_cancer_calcium.html#ixzz3ZCT65Fiv

Aleksandrowicz et al in Poland conclude that inadequacy of Mg (Magnesium) and antioxidants are important risk factors in predisposing to leukemias. Other researchers found that 46% of the patients admitted to an ICU (Intensive Care Unit) in a tertiary cancer center presented hypomagnesemia.

They concluded that the incidence of hypomagnesemia in critically ill cancer patients is high. In animal studies we find that Mg deficiency has caused lymphopoietic neoplasms in young rats. A study of rats surviving Mg deficiency sufficient to cause death in convulsions during early infancy in some, and cardiorenal lesions weeks later in others, disclosed that some of survivors had thymic nodules or lymphosarcoma.

One would not normally think that Magnesium (Mg) deficiency can paradoxically increase the risk of, or protect against cancer yet we will find that just as severe dehydration or asphyxiation can cause death, magnesium deficiency can directly lead to cancer. When you consider that over 300 enzymes and ion transport require magnesium and that its role in fatty acid and phospholipid acid metabolism affects permeability and stability of membranes, we can see that magnesiumdeficiency would lead to physiological decline in cells setting the stage for cancer. Anything that weakens cell physiology will lead to the infections that surround and penetrate tumor tissues. These infections are proving to be an integral part of cancer. Magnesium deficiency poses a direct threat to the health of our cells. Without sufficient amounts, our cells calcify and rot in. Breeding grounds for yeast and fungi colonies they become, invaders all too ready to strangle our life force and kill us.

Over 300 different enzymes systems rely upon magnesium to facilitate their catalytic action, including ATP metabolism, creatine-kinase activation, adenylate-cyclase, and sodium-potassium-ATPase.

It is known that carcinogenesis induces magnesium distribution disturbances, which cause magnesium mobilization through blood cells and magnesium depletion in non-neoplastic tissues. Magnesium deficiency seems to be carcinogenic, and in case of solid tumors, a high level of supplemented magnesium inhibits carcinogenesis. Both carcinogenesis and magnesium deficiency increase the plasma membrane permeability and fluidity. Scientists have in fact found out that there is much less Mg++ binding to membrane phospholipids of cancer cells, than to normal cell membranes.

Read Full Post »

Thyroid Function and Disorders

Writer and Curator: Larry H. Bernstein, MD, FCAP 

Normal thyroid function is maintained by endocrine interactions between the hypothalamus, anterior pituitary and thyroid gland [Matfin, 2009]. Iodide is transported across the basement membrane of the thyroid cells by an intrinsic membrane protein called the Na/I symporter (NIS). At the apical border, a second iodide transport protein called pendrin moves iodide into the colloid, where it is involved in hormono-genesis. Once inside the follicle, most of the iodide is oxidized by the enzyme thyroid peroxidase (TPO) in a reaction that facilitates combination with a tyrosine molecule to ultimately form thyroxine (T4) and triiodothyronine (T3). Thyroxine is the major thyroid hormone secreted into the circulation (90%, with T3 composing the other 10%). There is evidence that T3 is the active form of the hormone and that T4 is converted into T3 before it can act physiologically.

All of the major organs in the body are affected by altered levels of thyroid hormone. These actions are mainly mediated by T3. In the cell, T3 binds to a nuclear receptor, resulting in transcription of specific thyroid hormone response genes.

Maternal thyroid hormones are essential for neural development in zebrafish.

Marco A Campinho, João Saraiva, Claudia Florindo, Deborah M Power Molecular endocrinology (Baltimore, Md.) 05/2014;
http://dx.doi.org:/10.1210/me.2014-1032

ABSTRACT Teleost eggs contain an abundant store of maternal thyroid hormones (THs) and early in zebrafish embryonic development all the genes necessary for TH signalling are expressed. Nonetheless the function of THs in embryonic development remains elusive. To test the hypothesis that THs are fundamental for zebrafish embryonic development an MCT8 knockdown strategy was deployed to prevent maternal TH uptake. Absence of maternal THs did not affect early specification of the neural epithelia but profoundly modified later dorsal specification of the brain and spinal cord as well as specific neuron differentiation. Maternal THs acted upstream of pax2a, pax7 and pax8 genes but downstream of shha and fgf8a signalling. The lack of inhibitory spinal cord interneurons and increased motorneurons in the MCT8 morphants is consistent with their stiff axial body and impaired mobility. MCT8 mutations are associated with X-linked mental retardation in humans and the cellular and molecular consequences of MCT8 knockdown during embryonic development in zebrafish provides new insight into the potential role of THs in this condition.
Relationship between thyroid status and renal function in a general population of unselected outpatients

Giuseppe Lippi, Martina Montagnana, Giovanni Targher, Gian Luca Salvagno, Gian Cesare Guidi
Clin Biochem May 2008; 41(7–8): 625-627

When compared with euthyroid subjects, those with TSH < 0.2 mIU/L and > 2.5 mIU/L had increased and decreased estimated glomerular filtration rate (e-GFR), respectively. TSH levels were an independent predictor of e-GFR.

Serum Thyroid-Stimulating Hormone Measurement for Assessment of Thyroid Function and Disease

Douglas S. Ross
Endocr and Metab Clinics of N Am, Jun 2001; 30(2, 1): 245-264

Thyrotropin, or thyroid-stimulating hormone (TSH), is one of a family of glycoprotein hormones including luteinizing hormone (LH), follicle-stimulating hormone (FSH), and human chorionic gonadotropin (hCG) that share a common α-subunit and a unique β-subunit. Pituitary TSH regulates the secretion of the thyroid hormones T4 (thyroxine) and T3 (triiodothyronine). TSH secretion, in turn, is controlled through negative feedback by thyroid hormone on the pituitary thyrotrope. This relationship is negative log-linear. Small changes in serum free thyroid hormone concentrations result in large changes in serum TSH concentrations, and even subtle changes in thyroid hormone production are best assessed by measurement of serum TSH . Until the late 1980s, the detection limit of TSH assays was within the normal range, and these first-generation TSH assays were useful only for the detection of hypothyroidism. Free T4 measurements were primarily used for assessing thyroid function despite the technical difficulties in free thyroid hormone measurements owing to abnormal binding proteins, changes in binding protein concentrations, and the effects of drugs and illness on thyroid hormone binding. With the use of sensitive second- and third-generation TSH assays, TSH measurement has emerged as the single most useful test of thyroid function. It is widely and appropriately used as a screening test. Unfortunately, the trend has been to rely on TSH measurements alone for the assessment of complicated thyroid disease and patients undergoing treatment for thyroid dysfunction. This article focuses on the potential and real limitations of TSH measurement.
Correlation of creatinine with TSH levels in overt hypothyroidism — A requirement for monitoring of renal function in hypothyroid patients?

Vandana Saini, Amita Yadav, Megha Kataria Arora, Sarika Arora, Ritu Singh, Jayashree Bhattacharjee
Clin Biochem  Feb 2012; 45(3): 212-214

Highlights
► Increase serum creatinine levels in both subclinical and overt hypothyroidism. ► Creatinine levels progressively increase with increasing degree of hypothyroidism. ► Increase in creatinine correlated with TSH levels in overt hypothyroid subjects. ► Regular monitoring of renal function is required in hypothyroid patients.

Renal function is influenced by thyroid status. Therefore, this study was done to determine the relationship between renal function and different degrees of thyroid dysfunction.
Design and methods
Thyroid and kidney function tests were analyzed in 47 patients with overt (TSH ≥ 10.0 μIU/L) and 77 patients with subclinical hypothyroidism (TSH 6.0–9.9 μIU/L) in a cross-sectional study. These were compared with 120 age- and sex-matched euthyroid controls.
Results
Overt hypothyroid subjects showed significantly raised serum urea, creatinine and uric acid levels as compared to controls whereas subclinical hypothyroid patients showed significant increased levels of serum urea and creatinine levels. TSH showed significant positive correlation with creatinine and uric acid values and, fT4 had a negative correlation with uric acid in overt hypothyroidism.
Conclusion
Hypothyroid state is associated with significant derangement in biochemical parameters of renal function. Hence the renal function should be regularly monitored in hypothyroid patients.

  1. Ability of Serum Thyroid-Stimulating Hormone Levels to Reflect Peripheral and Central Thyroid Hormone Action Appropriately
  • Uncertainty Owing to Heterogeneity of T4 Deiodinases
  • Uncertainty Owing to Heterogeneity of T3 Receptors
  • Uncertainty Owing to Resetting of the Threshold for Negative Feedback
  1. Clinical Utility of Thyroid-Stimulating Hormone Measurement
  2. Screening for Thyroid Disease and Assessment of Patients Suspected of Having Thyroid Disease
  • Limitations of Thyroid-Stimulating Hormone Testing in Patients with Known Thyroid Disease Central Hypothyroidism
  • Thyrotoxicosis Owing to Inappropriate Thyroid-Stimulating Hormone Secretion
  • Monitoring Thyroid Hormone Therapy
  • Patients Treated for Hyperthyroidism
  1. The Pituitary-Thyroid Axis in Nonthyroidal Illness
  • Measurement of Thyroid-Stimulating Hormone
  • Drugs that Affect Serum Thyroid-Stimulating Hormone Concentrations

Investigations into the etiology of elevated serum T3 levels in protein-malnourished rats

Robert C. Smallridge, Allan R. Glass, Leonard Wartofsky, Keith R. Latham, Kenneth D. Burman
Metabolism, V June 1982; 31(6): 538-542

Thyroid function studies and the peripheral metabolism of thyroid hormone were examined in rats fed a low protein diet (9% casein) for 4–8 wk. Compared to animals fed a normal protein diet ad libitum, both the low protein rats and a pair-fed control group weighed less at the end of the study. However, serum total T3 levels were significantly higher only in the protein deficient rats. The elevated serum T3 was not explainable by enhanced peripheral T4 to T3 conversion, as there was no evidence of any change in hepatic or renal 5′-deiodinase activity when homogenates were examined for conversion of T4 to T3, reverse T3 to 3,3′-diiodothyronine, or 3′,5′-diiodothyronine to 3′-monoiodothyronine. Neither was there an effect on hepatic T3 receptor maximal binding capacity (204 ± 24 versus 168 ± 15 fmol/mg DNA control) or binding affinity (2.07 ± 0.38 versus 2.49 ± 0.24 × 10−10 M control). In two separate experiments the dialyzable fraction of T3 was significantly lower in the low protein group while free T3 concentrations were unchanged or reduced. In contrast, serum total and free T4 were either normal or reduced and dialyzable T4 was unaffected by protein deficiency. We conclude that while serum total T3 is elevated in rats chronically fed a low protein diet, this elevation is not due to enhanced T4 to T3 conversion. Rather, the increased T3 levels can be accounted for by a striking alteration in protein binding to T3. Moreover, the failure to demonstrate similar changes in serum total and dialyzable T4 suggests that in the rat, protein deficiency has different effects on binding to the two major thyroid hormones. Dietary induced changes in serum thyroid hormone binding must be kept in mind in nutrition studies in the rat.

Role of thyrotropin in metabolism of thyroid hormones in nonthyroidal tissues

Udaya M. Kabadi
Metabolism, Jun 2006; 55(6): 748-750

T4 conversion into T3 in peripheral tissues is the major source of circulating T3. However, the exact mechanism of this process is ill defined. Several in vitro studies have demonstrated that thyrotropin facilitates deiodination of T4 into T3 in liver and kidneys. However, there is a paucity of in vitro studies confirming this activity of thyrotropin. Therefore, this study was conducted to examine the influence of thyrotropin on thyroid hormone metabolism in nonthyroidal tissues. We assessed T4, T3, reverse T3 (rT3), and T3 resin uptake (T3RU) responses up to 12 hours at intervals of 4 hours in 6 thyroidectomized female mongrel dogs rendered euthyroid with LT4 replacement therapy before and after subcutaneous (SC) administration of bovine thyrotropin (5 U) on one day and normal saline (0.5 mL) on another in a randomized sequence between 08:00 and 09:00 am. Euthyroid state after LT4 replacement was confirmed before thyrotropin administration. Serum T4, T3, rT3, and T3RU all remained unaltered after SC administration of normal saline. No significant alteration was noted in serum T3RU values on SC administration of thyrotropin. However, serum T3 rose progressively reaching a peak at 12 hours with simultaneous declines being noted in both serum T4 and rT3 concentrations (P < .05 vs prethyrotropin values for all determinations). The changes after SC administration were significantly different (P < .001) in comparison to those noted on SC administration of normal saline. Thyrotropin may promote both the conversion of T4 to T3 and metabolism of rT3 into T2 in nonthyroidal tissues via enhancement of the same monodeionase.

Effects of growth hormone administration on fuel oxidation and thyroid function in normal man

Jens Møller, Jens O.L. Jørgensen, Niels Møller, Jens S. Christiansen, Jørgen Weeke
Metabolism, Jul 1992;  41(7): 728-731

In a randomized, double-blind, placebo-controlled, cross-over study, we examined the effects of 14 days of growth hormone (GH) administration (12 IU/d subcutaneously) on energy expenditure (EE), respiratory exchange ratio (RER), and thyroid function in 14 normal adults of normal weight (eight men and six women). EE (kcal/24 h) was significantly elevated after GH administration (2,073 ± 392, [GH], 1,900 ± 310, [placebo], P = .01). RER was significantly lowered during GH administration (0.73 ± 0.04 v 0.78 ± 0.06, P = .02), reflecting increased oxidation of lipids. Total triiodothyronine (TT3) (nmol/L) and free T3 (FT3) (pmol/L) increased significantly during GH (TT3: 1.73 ± 0.06 [GH], 1.48 ± 0.08 [placebo], P = .01; FT3: 6.19 ± 0.56 [GH], 5.49 ± 0.56 [placebo], P = .01). Concomitantly, an insignificant decrease in reverse T3 (rT3) (nmol/L) was observed (0.07 ± 0.01 [GH], 0.15 ± 0.01 [placebo], P = .08). GH caused a highly significant increase in T3/thyroxine (T4 (×100) ratio (1.84 ± 0.12 [GH], 1.37 ± 0.06 [placebo]). Serum thyrotropin (TSH) was not significantly changed by GH. No changes in total thyroxine (TT4) (nmol/L) (98 ± 6 [GH], 111 ± 8 [placebo], P = .40) and free thyroxine (FT4) (pmol/L) (17.4 ± 1.3 [GH], 18.6 ± 1.1 [placebo], P = .37) after 14 days of GH administration were observed. In conclusion, 2 weeks of GH administration increases EE and lipidoxidation. This finding may partly be mediated by an increase in peripheral T4 to T3 conversion.

Studies on the deiodination of thyroid hormones in Xenopus laevis tadpoles

Helen Robinson, Valerie Anne Galton
Gen Compar Endocr, Sept 1976; 30(1): 83-90

Liver and tail tissues from Xenopus laevis tadpoles possess deiodinating systems capable of degrading both thyroxine (T4) and 3,5,3′-triiodothyronine (T3). Deiodinating activity in liver remains at a constant level throughout late development and metamorphosis with the exception of a transient increase at stage 59, the onset of metamorphosis. Tail activity remains constant during development but rises sharply during metamorphosis when the tail is undergoing regression. In contrast to these findings on spontaneously metamorphosing tadpoles, tail tips induced to regress in vitro do not exhibit any rise in deiodinating activity, even when the tail tips are undergoing extensive autolysis. These results indicate that, while a rise in deiodinating activity may coincide temporarily with hormone action during metamorphosis, the two phenomena may be separated. The deiodinating activity present in tadpole tissues appears to be enzymic and possesses properties characteristic of peroxidase activity. The reaction catalyzed by this mechanism does not appear to involve monodeiodination and hence cannot be considered a mechanism for the peripheral conversion of T4 to T3.

Mechanisms governing the relative proportions of thyroxine and 3,5,3′-triiodothyronine in thyroid secretion

Peter Laurberg
Metabolism, Apr 1984; 33(4): 379-392

In subjects with normal thyroid function only a minor part of circulating 3,5,3′-triiodothyronine (T3) originates directly from the thyroid; the majority is produced in the peripheral tissues by deiodination of thyroxine (T4). However, T3 of thyroidal origin constitutes a relatively high fraction of the total T3 produced in many patients with thyroid hyperfunction or hypofunction. Such a relatively high T3 content in the secretion of the thyroid could be caused by a low T4T3 ratio in thyroglobulin. Severe iodine deficiency is a well-known inducer of a low T4T3 ratio, but a low T4T3 ratio can also be produced independent of the iodine content. This is seen in in vitro studies of thyroglobulin iodination when small amounts of DIT are added to the incubation mixture and in vivo in TSH-treated animals and in patients with Graves’ disease. Another mechanism for high thyroidal secretion of T3 could be an enhanced fractional deiodination of T4 to T3 in the thyroid. In vitro thyroid perfusion studies have shown that the T3 content of thyroid secretions is higher than would be expected from the T4T3 ratio of thyroid hydrolysate and that the major mechanism is deiodination of T4 to T3. Thyroxine deiodinases are also present in the human thyroid, and the amount of T4 deiodinase is enhanced in the thyroids from patients with medically treated Graves’ disease and in the hyperstimulated thyroids of rats. Other factors of possible importance for the mixture of T3 and T4 secreted by the thyroid are a relatively faster liberation of T3 than of T4 from thyroglobulin during partial hydrolysis (this faster release of T3 is probably the mechanism behind the more “rapid” secretion of T3 than of T4), or some kind of thyroid heterogeneity leading to pinocytosis and hydrolysis of thyroglobulin with a lower T4T3 ratio than that of average thyroglobulin.

Starvation-induced alterations of circulating thyroid hormone concentrations in man

Thomas J. Merimee, E.S. Fineberg
Metabolism Jan 1976; 25(1): 79-83

Serum concentrations of triiodothyronine (T3), thyroxine (T4), and TSH were examined in seven men and seven women of normal weight during a 60-hr fast. Similar studies were conducted in two women who received daily for 1 mo before and during a similar fast, 0.4 mg and 0.5 mg of l-thyroxine.
The serum concentrations of T3 decreased in each of the untreated normal subjects (sign test of significance, p < 0.001). The mean control concentration of T3 in women was 152 ± 9 ng100 ml (X ± SEM); after 24 hr of fasting, 131 ± 31 ng100 ml; and at the termination of the fast, 90 ± 15 ng100 ml. The latter value differed from the control value with a p value of < 0.01. Similar changes of T3 concentration occurred in men (mean basal T = 160 ± 11 ng100 ml; mean at termination of fast = 87 ± 16 ng100 ml). The range of decrease for T3 in all subjects varied from 24% to 55%.
The mean T4 concentration at the beginning of the fast was  6.9 ± 0.9, and at the termination of the fast, 7.5 ± 0.6 (p = NS). TSH concentrations remained unchanged (Control, 3.8 ± 0.45 μU/ml; at 60 hr, 4.0 ± 0.26 μU/ml, p = NS).
Studies in two women who received, before and during a fast, T4, indicate that a decreased peripheral conversion of T4 to T3 is the most likely mechanism responsible for this change.

Effect of estrogens on thyroid function. II. Alterations in plasma thyroid hormone levels and their metabolism

Ramesh C. Sawhney, Indra Rastogi, Gopal K. Rastogi
Metabolism Mar 1978; 27(3): 279-288

The circulating levels of total triiodothyronine (TT3), thyroxine (TT4, and T4-bbinding globulin (TBG) and the kinetics of T3 and T4 were studied in five menstruating rhesus monkeys before, during, and after prolonged treatment with estradiol monobenzoate (E2B, 50 μg/kg body weight/day subcutaneously). A significant increase over pretreatment (p < 0.01) plasma TT3, TT4, and TBG was recorded on day 6 of E2B therapy. A further significant stepwise increase in these parameters was noted up to day 19 of E2B, when the levels plateaued for the rest of the period of E2B treatment. Two weeks after discontinuation of E2B, plasma TT3, TT4, and TBG had returned to the pretreatment range and remained so up to 40 days of observation. Although the percent free T3 and percent free T4 were significantly decreased (p < 0.01) during E2B therapy, the absolute concentrations of free T3 and free T4 were not altered. After prolonged E2B treatment the metabolic clearance rate, distribution space, and production rate (PR) of both T3 and T4 were decreased (p < 0.01). The extrathyroidal T4 pool (ETT4P) was significantly increased (p < 0.01), whereas ETT3P did not show any significant alterations (p > 0.05). The decreased PR of T4 might have been due to a direct inhibitory effect of E2B on the thyroid, whereas the decrease in PR of T3 might have been due to either decreased conversion of T4 to T3, to decreased secretion by the thyroid, or both.
Zebrafish as a model to study peripheral thyroid hormone metabolism in vertebrate development

Marjolein Heijlen, Anne M. Houbrechts, Veerle M. Darras
Gen Compar Endocr 1 Jul 2013; 188: 289-296

To unravel the role of thyroid hormones (THs) in vertebrate development it is important to have suitable animal models to study the mechanisms regulating TH availability and activity. Zebrafish (Danio rerio), with its rapidly and externally developing transparent embryo has been a widely used model in developmental biology for some time. To date many of the components of the zebrafish thyroid axis have been identified, including the TH transporters MCT8, MCT10 and OATP1C1, the deiodinases D1, D2 and D3, and the receptors TRα and TRβ. Their structure and function closely resemble those of higher vertebrates. Interestingly, due to a whole genome duplication in the early evolution of ray-finned fishes, zebrafish possess two genes for D3 (dio3 and dio3a) and for TRα (thraa and thrab). Transcripts of all identified genes are present during embryonic development and several of them show dynamic spatio-temporal distribution patterns. Transient morpholino-knockdown of D2, D3 or MCT8 expression clearly disturbs embryonic development, confirming the importance of each of these regulators during early life stages. The recently available tools for targeted stable gene knockout will further increase the value of zebrafish to study the role of peripheral TH metabolism in pre- and post-hatch/post-natal vertebrate development.

The consequences of inappropriate treatment because of failure to recognize the syndrome of pituitary and peripheral tissue resistance to thyroid hormone

Samuel Refetoff, Angel Salazar, Terry J. Smith, Neal H. Scherberg
Metabolism  Aug 1983; 32(8); 822-834

Since the description of the syndrome of global (peripheral tissues and pituitary) resistance to thyroid hormone, new cases are being recognized with increasing frequency. The patient described herein had a markedly elevated serum TSH concentration of 260 μU/mL at the time of diagnosis. Studies suggest that elevations of serum TSH levels in this and other patients with the syndrome are most likely iatrogenic in origin. The patient was 312 years old when a goiter and a high serum T4 concentration were detected. Despite subtotal thyroidectomy, antithyroid drugs were required to maintain her T4 level in the normal range. She was referred at age 1112 years because of recurrent goiter. Her parents and five older siblings had normal thyroid function. Off therapy, her serum T4 level was 14.9 μg/dL, FT4I was 17.0, T3 was 362 ng/dL, TSH was 260 μU/mL, and antibodies were negative. There were no signs of thyrotoxicosis, her bone age was 7 years, her growth was stunted (third percentile), her intellectual quotient (IQ) was 67, and there was a 30–50 dB sensorineural hearing loss. The presence of a pituitary adenoma was ruled out. Her TSH had normal bioreactivity and rose to 540 μU/mL in response to TRH. Triiodothyronine was given in incremental doses of 50, 100, 200, and 400 μg/d over 28 days. The log concentrations of serum TSH showed an inverse linear correlation with serum T3. While receiving the highest dose of T3, on which the level of serum T3 ranged from 1400 to 2500 ng/dL, the TSH response to TRH normalized (basal 4.2 and peak 20 μU/mL), as did the high levels of serum cholesterol, carotene, and T4. Her BMR rose from +5 to +22%, her IQ rose to 77, and she gained weight without an increase in caloric intake. Only minimal changes were observed in levels of urinary cAMP, hydroxyproline, magnesium, and nitrogen. All values, with the exception of the weight gain, returned to baseline 2 months after T3 treatment was discontinued. The TSH level was suppressed by l-dopa and by prednisone. Long-term therapy with equivalent doses of T4 (from 300 to 1000 μg/d) produced a growth of 3 cm during the initial 6 weeks, 10.5 cm over the ensuring year (above the 10th percentile), and regression of goiter without thyrotoxicosis. The patient exhibited resistance to thyroid hormone in pituitary and peripheral tissues. The optimal dose of T4 replacement could be predicted by studying tissue responses to incremental doses of T3. The marked elevation in serum TSH concentration, stunted growth, and laboratory evidence of hypothyroidism were due to the limited thyroidal reserve caused by thyroidectomy. All patients with an impaired ability to compensate for the defect as a result of inappropriate treatment should be given thyroid hormone in amounts short of producing catabolic effects. Such a dose is expected to normalize the basal serum TSH concentration and its response to TRH.

Solving the mystery of iodine uptake

Valda Vinson
Science 20 Jun 2014; 344(6190), p. 1355
http://dx.doi.org:/10.1126/science.344.6190.1355-a

The thyroid gland produces iodine-containing hormones that regulate metabolism. The cell membrane protein NIS (sodium/iodine symporter) transports iodine into thyroid cells, but because iodine concentrations outside of the cell are so low, how it does so is a mystery. The key? Moving two sodium ions along with the iodine ion, Nicola et al found. NIS also does not bind sodium very tightly, but the high concentrations of sodium outside the cell allow one sodium ion to bind. This binding increases the affinity of NIS for a second sodium ion and also for iodine. With the three ions bound, NIS changes its conformation so that it opens to the inside of the cell, where the sodium concentration is low enough for NIS to release its sodium ions. When the sodium goes away, so does NIS’s affinity for iodine, leading NIS to release it.

Unliganded Thyroid Hormone Receptor α Regulates Developmental Timing via Gene Repression in Xenopus tropicalis

Jinyoung Choi, Ken-ichi T. Suzuki, Tetsushi Sakuma, Leena Shewade, Takashi Yamamoto, and Daniel R. Buchholz
Endocr Feb 2015; 156(2): 735–744 http://dx.doi.org:/10.1210/en.2014-1554

Thyroid hormone (TH) receptor (TR) expression begins early in development in all vertebrates when circulating TH levels are absent or minimal, yet few developmental roles for unliganded TRs have been established. Unliganded TRs are expected to repress TH-response genes, increase tissue responsivity to TH, and regulate the timing of developmental events. Here we examined the role of unliganded TRα in gene repression and development in Xenopus tropicalis. We used transcription activator-like effector nuclease gene disruption technology to generate founder animals with mutations in the TRα gene and bred them to produce F1 offspring with a normal phenotype and a mutant phenotype, characterized by precocious hind limb development. Offspring with a normal phenotype had zero or one disrupted TRα alleles , and tadpoles with the mutant hind limb phenotype had two truncated TRα alleles with frame shift mutations between the two zinc fingers followed by 40–50 mutant amino acids and then an out-of-frame stop codon. We examined TH-response gene expression and early larval development with and without exogenous TH in F1 offspring. As hypothesized, mutant phenotype tadpoles had increased expression of TH-response genes in the absence of TH and impaired induction of these same genes after exogenous TH treatment, compared with normal phenotype animals. Also, mutant hind limb phenotype animals had reduced hind limb and gill responsivity to exogenous TH. Similar results in methimazole-treated tadpoles showed that increased TH-response gene expression and precocious development were not due to early production of TH. These results indicate that unliganded TRα delays developmental progression by repressing TH-response genes.
The discovery of thyroid replacement therapy. Part 2: The critical 19th century
Conceptualizing the link between the thyroid and myxoedema

Stefan Slater
R Soc Med 2011; 104: 59–63. http://dx.doi.org:/10.1258/jrsm.2010.10k051

Sir William Withey Gull (1816–1890)

Frederik Ruysch, anatomist in Leyden around 1690, adopted, according to Albrecht von Haller in 1766, the opinion that a peculiar fluid was elaborated in the gland and poured into the veins’. The 19th century thus began with thyroidology at best in embryo; but during that century endocrinology was born and the thyroid was its standard bearer. In 1836, Thomas Wilkinson King of Guys Hospital, regarded by some as the ‘Father of Endocrinology’, anticipated on the basis of observation and experiment the internal secretion of the thyroid. In a meticulous paper on its anatomy: he wrote of the thyroid gland that ‘its absorbent vessels carry its peculiar secretion to the great veins of the body’. This language is almost identical to that of Ruysch and Haller more than a century earlier. The idea was prompted by the thyroid’s disproportionately large vascular supply in the absence of any evident mechanical or other local function and also at what he described as its ‘peculiar’ fluid. King notes that his view ‘has been indirectly surmised by Morgagni [probably in 1761] and others’.
In 1850, at a meeting of the Royal Medical and Chirurgical Society of London, chaired by Thomas Addison, Thomas Blizzard Curling, surgeon at the London Hospital, provided a clear clinicopathological correlate in a paper entitled ‘Two cases of absence of the thyroid body and symmetrical swellings of fat tissue at the sides of the neck, connected with defective cerebral development’.  Postmortem examination in each revealed no trace of thyroid tissue and that the swellings consisted only of fat.  Curling’s important observation was not pursued until 1871 when, at another meeting of the Society, Curling himself then in the chair, Charles Hilton Fagge, a physician at Guy’s Hospital, presented a paper on sporadic cretinism. He described four living cases and noted that none of them had a goiter and that one had been well up to the age of eight and, although now physically cretinous at age 16, she remained very intelligent. He referred to Curling’s paper and reached the same conclusion that the ‘healthy thyroid body is capable of exerting a counteracting influence [on cretinism]’.
Two years later, in 1873, Fagge’s senior colleague at Guy’s, Sir William Withey Gull, presented before the Clinical Society of London two of the five cases he had seen of what he called ‘A Cretinoid State supervening in Adult Life in Women’. He described their cretin-like appearance, drawing particular attention to the broad and thick tongue and the guttural voice and its pronunciation ‘as if the tongue were too large for the mouth’. He acknowledged his remarks were tentative, hence, he said, his use of the word ‘cretinoid’, but he had no doubt this was a ‘substantive’ condition and not one of cardiac or renal origin.
Gull was an interesting personality with apparently a remarkable presence, resembling Napoleon in face, form and manner (Figure). In the 1970s, 80 years after his death in 1890, he was the subject of a theory, quickly discredited, that he had been ‘Jack the Ripper’, the killer in the still unsolved murders and mutilations of at least five Whitechapel prostitutes in 1888. He figured in the 1988 TV film series, Jack the Ripper, starring Michael Caine as the detective. Gull is credited with the first description of hypothyroidism in adults and his paper was important in defining a recognizable clinical syndrome.
Then, in 1877, William Miller Ord, read his paper before the Royal Medical and Chirurgical Society of London and proposed the term ‘myxoedema’ for the adult condition. He described the non-pitting, ‘mucous edema’.   He also presented an engaging theory to explain the lethargy, inertia and slow responses associated with the disease. He suggested that these might result from the sheathing and insulation of the body in a ‘jelly-like’, mucin-laden integument that interfered with sensory perceptions and stimulation. Six years later, he chaired the committee set up by the Clinical Society of London to investigate the whole matter. He also later undertook some of the earliest metabolic studies of the effects of treating myxoedema with thyroid extract, showing the rapid weight loss and rise in temperature and in urinary volume and nitrogen excretion that occurred.
The key papers, which advanced these English authors observations, were those of the Swiss surgeons, Jaques-Louis Reverdin in Geneva and Emil Theodor Kocher in Bern, Kocher later receiving the Nobel Prize for his work on the thyroid. How fitting it is that it should be two Swiss doctors whose practices unlocked an understanding of the importance of the thyroid. For they each identified the late effects of total ablation (extirpation) of goiters. they

noted the great similarity of Gull’s and Ord’s myxoedema cases with their affected postoperative patients, referring to the comparison as a ‘rapprochement complet’, clearly making the connection. They acknowledged Gull’s primacy in describing the clinical manifestations and Ord’s ‘christening’ the condition ‘myxoedema’, and proposed that surgical cases be known as ‘myxoedème opératoire’. In light of his findings in 1882, Reverdin thereafter sought to conserve a part of the gland during thyroidectomy for goiter, speculating that its complete removal may have been responsible for these late effects. He had noticed that no such problems followed a just unilateral lobectomy. Kocher called the disease picture in his affected cases ‘cachexia strumipriva’ – literally, a bad condition due to the removal of a struma (goiter) without reference to the earlier work of Reverdin. Halsted noted in his monumental review of goiter surgery: ‘It is interesting to follow the argumentation of a mind so exceptionally keen and sane as Kocher’s in its futile efforts to explain insufficiently illuminated phenomena’. In reading Kocher’s 1909 Nobel Prize Lecture (in English translation), one gets the impression that Kocher was aware in 1883 of Gull’s and Ord’s reports, despite not referring to them, and he dismisses Reverdin’s contribution.
There ensued a competition over the contribution to the thyroid discovery.  When post-thyroidectomy myxedema wsas brought to the attention of Kocher, he agreed it was analogous to his cases of cachexia strumipriva. It is also obvious that Kocher, like many surgeons of the time, cannot have engaged in routine postoperative outpatient follow-up, for otherwise the ensuing problems in his goiter-operated patients would have been detected years earlier. In respect of this key moment in the history of the thyroid, Reverdin could be said to hold the intellectual property. The thought has been expressed that perhaps he should have shared the 1909 Nobel Prize with Kocher.
The Emerging Roles of Thyroglobulin

Yuqian Luo, Yuko Ishido, Naoki Hiroi, Norihisa Ishii, and Koichi Suzuki
Adv in Endocr 2014, Article ID 189194, 7 pp http://dx.doi.org/10.1155/2014/189194

Thyroglobulin (Tg), the most important and abundant protein in thyroid follicles, is well known for its essential role in thyroid hormone synthesis. In addition to its conventional role as the precursor of thyroid hormones, we have uncovered a novel function of Tg as an endogenous regulator of follicular function over the past decade. The newly discovered negative feedback effect of Tg on follicular function observed in the rat and human thyroid provides an alternative explanation for the observation of follicle heterogeneity. Given the essential role of the regulatory effects of Tg, we consider that dysregulation of normal Tg function is associated with multiple human thyroid diseases including autoimmune thyroid disease and thyroid cancer. Additionally, extrathyroid Tg may serve a regulatory function in other organs. Further exploration of Tg action, especially at the molecular level, is needed to obtain a better understanding of both the physiological and pathological roles of Tg.

The Surgical Management of Thyroid Cancer

Sara A. Morrison, Hyunsuk Suh, and Richard A. Hodin
Rambam Maimonides Med J 2014; 5(2):e0008. http://dx.doi.org:/10.5041/RMMJ.10142

There are approximately 63,000 reported cases of thyroid carcinoma annually in the United States, representing roughly 4% of all documented malignancies.1 Diagnosis typically stems from work-up of a thyroid nodule. Data from the Framingham study suggests that palpable thyroid nodules are present in 4% of the US population,2 but non-palpable nodules may exist in up to 67% of the population. Such nodules are often found incidentally secondary to the rising use of imaging modalities in medical settings. The large majority of thyroid nodules are benign, with an overall reported risk of malignancy from 5% to 15%.
Thyroid cancer has been increasing in incidence, with the number of reported cases in the US rising by 25% over the last 3 years. With growing technological advances in the field and improved contributions of diagnostics, surgical decision-making and operative planning have taken on new challenges. Herein, we review the current clinical practice recommendations and active areas of surgical controversy, reflective of the most recently published professional consensus guidelines and a systematic review of the literature.
The use of FNA in current clinical practice has resulted in post-surgical pathology findings of malignancy in over 50% of specimens.7 The Bethesda System for Reporting Thyroid Cytopathology (TBSRTC) was developed in order to allow pathologists among varying institutions to communicate results to clinical care-takers with widely under-stood descriptors. Results of FNA biopsies are broken down into the following categories with the corresponding risks of malignancy: non-diagnostic or unsatisfactory (1%–4%), benign (0%–3%), atypia of undetermined significance or follicular lesion of undetermined significance (AUS/FLUS; 5%–15%), follicular neoplasm or suspicious for a follicular neoplasm (FN/sFN; 15%–30%), suspicious for malignancy (60%–75%), and malignant (97%–99%).
Mutational Panels.
AsuragenmiR Inform (Austin, TX, USA) mutation analysis assay and Thyroid Cancer Mutation Panel by Quest Diagnostics (Madison, NJ, USA) are the two main commercially available mutational tests which test for known genetic alterations such as BRAF, RAS, RET/PTC, and PAX8/PPAR. These mutational panels are highly specific for malignancy; however, due to the low overall frequency of these mutations in thyroid cancers, negative results do not rule out cancer. Therefore, mutational panel tests are considered a “rule-in” test. If a preoperative mutational test is positive, the nodule should be considered malignant, and total thyroidectomy should be recommended.
Gene Expression Profiling.
The most widely known gene expression profiling test is Afirma Gene Expression Classifier (Veracyte, San Francisco, CA, USA), and, with its recent clinical validation by Alexander et al., Afirma is already being utilized in many clinical settings. The Afirma Gene Expression Classifier (GEC) is an RNA-based assay that utilizes FNA samples to evaluate 167 molecular genes associated with benign nodules based on their proprietary algorithm. Unlike the mutational panel testing, Afirma testing is considered a “rule-out” test since the test has a high negative predictive value in distinguishing benign nodules. However, a positive result reported as “suspicious” carries only 38% risk of malignancy.
In all, these molecular tests should be utilized judiciously and should be considered as a complementary diagnostic tool in the management of thyroid nodules. In the future, molecular testing could become more cost-effective and accurate as a diagnostic tool while providing prognostic and therapeutic information.
Papillary Thyroid Cancer.
Total thyroidectomy is the gold standard for patients with a preoperative diagnosis of papillary thyroid cancer when the nodule is greater than 1 cm in size. Completion thyroidectomy is indicated in patients who have undergone prior lobectomy and are found on final pathology to have papillary thyroid cancer that is larger than 1 cm. The completion thyroidectomy should generally be performed within 6 months of the original procedure in order to minimize the risk of lymph node metastasis.
Involvement of cervical lymph nodes in papillary thyroid cancer is frequent, reported to occur in up to 50% of patients. The role of neck dissection at the time of total thyroidectomy is somewhat controversial, however, since most of the nodal involvement is microscopic and does not affect overall survival. It is generally agreed upon that a therapeutic neck dissection should be pursued in the setting of well-differentiated thyroid cancer patients with clinically positive lymph nodes, whether in the central or lateral neck compartments. Prophylactic neck dissection is not done for follicular thyroid cancer, as the rates of lymph node metastasis are typically less than 10%.
Medullary thyroid cancer (MTC) comprises 4% of all thyroid malignancies. The majority of cases are sporadic in nature; approximately 20%–25% represent familiar/hereditary syndromes. Diagnosis is commonly made by FNA biopsy with specific staining for the presence of calcitonin in the tissue specimen. All patients with a diagnosis of medullary thyroid cancer must be evaluated for multiple endocrine neoplasia (MEN) 2 and be ruled out for the synchronous presence of pheochromocytoma prior to scheduling thyroid surgery.
Effects of Dose Level of Anti-thyroid Drug Carbimazole on Thermoregulation and Blood Constituents in Male Rabbits (Oryctolagus cuniculus)

Intisar H. Saeed, Abdalla M. Abdelatif and Mohamed E. Elnageeb
Adv in Research 2014; 2(3): 129-144. Article no. AIR.2014.002

Carbimazole (CBZ) is an anti-thyroid drug commonly used in the treatment of hyperthyroidism. The objective of this study was to evaluate the effects of dose level of CBZ on thermoregulation and blood constituents in mature male rabbits. Twenty animals were assigned to 4 groups (A, B, C, D) of 5 each. Group A served as control and treated animals in groups B,C,D, received daily orally CBZ doses of 10, 15 and 20 mg/animal for 3 weeks, respectively.
The values of rectal temperature (Tr,), respiration rate (RR) and heart rate (HR) decreased in treated rabbits and the mean values of HR decreased with increase in the dose level of CBZ. The packed cell volume (PCV),  Hb concentration and total leukocyte count (TLC) were lower in CBZ treated rabbits. Serum levels of total protein and globulins increased and serum albumin level decreased in treated groups of rabbits. Serum urea level was lower in CBZ treated groups and there was an increase in serum urea level with increase in CBZ dose level. Serum cholesterol level was higher in treated groups and there was an increase in serum cholesterol level with increase in CBZ dose level. Plasma glucose level decreased significantly in CBZ treated groups compared with the control and the mean values decreased with increase in the dose level of CBZ. The results indicate that the responses of basic physiological parameters were almost dose dependent in the range adopted in this study.
Phosphatase Inhibitor Calyculin A Activates TRPC2 Channels in Thyroid FRTL-5 Cells

Pramod Sukumaran, MY Asghar, C Löf, T Viitanen, and Kid Törnquist
Calcium Signaling Jun 2014; 1(2)  http://www.researchpub.org/journal/cs/cs.html

We have previously shown that rat thyroid FRTL-5 cells express a calcium entry pathway regulated by a phosphatase. The nature of the calcium entry pathway is presently unknown. We have also shown that FRTL-5 cells express only the TRPC2 channel of the TRPC family of cation channels. In the present investigation we show, using pharmacological inhibitors, the measurement of sodium and calcium entry, stable TRPC2 knock-down cells, and transfection with a non-conducting form of TRPC2, that the calcium entry pathway regulated by a phosphatase is, in fact, the TRPC2 channel. Our data thus point to a novel mechanism by which the TRPC2 channels can be regulated.

Thyroxine Uptake by Perfused Rat Liver
No Evidence for Facilitation by Five Different Thyroxine-binding Proteins

Carl M. Mendel and Richard A. Weisiger
J. Clin. Invest.  1990; 86: 1840-1847

For each of the five protein-hormone complexes studied, the rate of hepatic uptake of T4 (measured under conditions expected to result in dissociation-limited uptake) closely approximated the rate of spontaneous dissociation of the protein-hormone complex within the hepatic sinusoids. These findings indicate an absence of special cellular mechanisms that facilitate the hepatic uptake of T4 from its plasma binding proteins, and support the view that uptake occurs from the free T4 pool after spontaneous dissociation of T4 from its binding proteins.
Thyroxine Transport and Distribution in Nagase Analbuminemic Rats

Carl M. Mendel, RR Cavalieri, LA Gavin, T Pettersson, and M Inoue
J. Clin. Invest. 1989; 83: 143-148

The postulate that thyroxine (T4) in plasma enters tissues by protein-mediated transport or enhanced dissociation from plasma-binding proteins leads to the conclusion that almost all T4 uptake by tissues in the rat occurs via the pool of albumin bound T4 (Pardridge, W. M., B. N. Premachandra, and G. Fierer. 1985. Am. J. Physiol. 248:G545-G550).
To directly test this postulate, and to test more generally whether albumin might play a special role in T4 transport in the rat, we performed in vivo kinetics studies in six Nagase analbuminemic rats and in six control rats, all of whom had similar serum T4 concentrations and percent free T4 values.
Evaluation of the plasma disappearance curves of simultaneously injected 125I-T4 and I31I-albumin indicated that the flux of T4 from the extracellular compartment into the rapidly exchangeable intracellular compartment was similar in the analbuminemic rats (51±21 ng/min, mean±SD) and in the control rats (54±15 ng/min), as was the size of the rapidly exchangeable intracellular pool of T4 (1.13±0.53 vs. 1.22±036 Mg). This latter finding was confirmed by direct analysis of tissue samples (liver, kidney, and brain). We also performed in vitro kinetics studies using the isolated perfused rat liver. The single-pass fractional extraction by normal rat liver of T4 in pooled analbuminemic rat serum was indistinguishable from that of T4 in pooled control rat serum (10.9±3.3%, n = 3, vs. 11.4±3.4%). When > 98% of the albumin was removed from normal rat serum by chromatography with Affi-Gel blue, the single-pass fractional extraction of T4 (measured by a bolus injection method) did not change (16.3±2.1%, n = 5, vs. 15.2±2.5%). These data provide the first valid experimental test of the enhanced

dissociation hypothesis and indicate that there is no special, substantive role for albumin in T4 transport in the rat.
Influence of thyroid receptors on breast cancer cell proliferation

  1. Conde, R. Paniagua, J. Zamora, M. J. Blanquez, B. Fraile, A. Ruiz & M. I. Arenas
    Ann Oncol 2005; http://dx.doi.org:/10.1093/annonc/mdj040

Background: The involvement of thyroid hormones in the development and differentiation of normal breast tissue has been established. However, the association between breast cancer and these hormones is controversial. Therefore, the objective of the present study was to determine the protein expression pattern of thyroid hormone receptors in different human breast pathologies and to evaluate their possible relationship with cellular proliferation.
Patients and methods: The presence of thyroid hormone receptors was evaluated by immunohistochemistry and western blot analysis in 84 breast samples that included 12 cases of benign proliferative diseases, 20 carcinomas in situ and 52 infiltrative carcinomas.
Results: TR-α was detected in the nuclei of epithelial cells from normal breast ducts and acini, while in any pathological type this receptor was located in the cytoplasm. However, TR-b presented a nuclear location in benign proliferative diseases and carcinomas in situ and a cytoplasmatic location in normal breast and infiltrative carcinomas. The highest proliferation index was observed in carcinomas in situ, although in infiltrative carcinomas an inverse correlation between this index and the TR-α expression was encountered.
Conclusions: The results of this study reveal substantial changes in the expression profile of thyroid hormone.
Zebrafish as a model for monocarboxyl transporter 8-deficiency

GD Vatine, D Zada, T Lerer-Goldshtein, A Tovin, G Malkinson, K Yaniv and L Appelbaum
J Biol Chem Nov 2012; Manuscript M112.413831
http://dx.doi.org:/10.1074/jbc.M112.413831

Background: Mutations in the thyroid hormone transporter MCT8 are associated with psychomotor retardation AHDS.
Results: In zebrafish, as in humans, mct8 is expressed primarily in the nervous system. Elimination of MCT8 causes severe neural impairment.
Conclusion: MCT8 is a crucial regulator during zebrafish embryonic development. Significance: Establishment of the first vertebrate model for MCT8-deficiency, which exhibits a neurological phenotype.
Unusual Ratio between Free Thyroxine and Free Triiodothyronine in a Long-Lived Mole-Rat Species with Bimodal Ageing

Yoshiyuki Henning, Christiane Vole, Sabine Begall, Martin Bens, et al.
PlusOne Nov 2014; 9(11),e113698. http://dx.doi.org:/10.1371/journal.pone.0113698

Ansell’s mole-rats (Fukomys anselli) are subterranean, long-lived rodents, which live in eusocial families, where the maximum lifespan of breeders is twice as long as that of non-breeders. Their metabolic rate is significantly lower than expected based on allometry, and their retinae show a high density of S-cone opsins. Both features may indicate naturally low thyroid hormone levels.
In the present study, we sequenced several major components of the thyroid hormone pathways and analyzed free and total thyroxine and triiodothyronine in serum samples of breeding and non-breeding F. anselli to examine whether
a) their thyroid hormone system shows any peculiarities on the genetic level,
b) these animals have lower hormone levels compared to euthyroid rodents (rats and guinea pigs), and
c) reproductive status, lifespan and free hormone levels are correlated.
Genetic analyses confirmed that Ansell’s mole-rats have a conserved thyroid hormone system as known from other mammalian species. Interspecific comparisons revealed that free thyroxine levels of F. anselli were about ten times lower than of guinea pigs and rats, whereas the free triiodothyronine levels, the main biologically active form, did not differ significantly amongst species. The resulting fT4:fT3 ratio is unusual for a mammal and potentially represents a case of natural hypothyroxinemia.
Comparisons with total thyroxine levels suggest that mole-rats seem to possess two distinct mechanisms that work hand in hand to downregulate fT4 levels reliably. We could not find any correlation between free hormone levels and reproductive status, gender or weight. Free thyroxine may slightly increase with age, based on subsignificant evidence. Hence, thyroid hormones do not seem to explain the different ageing rates of breeders and nonbreeders. Further research is required to investigate the regulatory mechanisms responsible for the unusual proportion of free thyroxine and free triiodothyronine.
Transthyretin Regulates Thyroid Hormone Levels in the Choroid Plexus, But Not in  the Brain Parenchyma: Study in a Transthyretin-Null Mouse Model

JA Palha, R Fernandes, GM De Escobar, V Episkopou, M Gottesman, and MJ Saraiva
Endocr 2000; 141(9): 3267–3272.

Transthyretin (TTR) is the major T4-binding protein in rodents. Using a TTR-null mouse model we asked the following questions.
1) Do other T4 binding moieties replace TTR in the cerebrospinal fluid (CSF)?
2) Are the low whole brain total T4 levels found in this mouse model associated with hypothyroidism, e.g. increased 59-deiodinase type 2 (D2) activity and RC3-neurogranin messenger RNA levels?
3) Which brain regions account for the decreased total whole brain T4 levels?
4) Are there changes in T3 levels in the brain?
Our results show the following.
1) No other T4-binding protein replaces TTR in the CSF of the TTR-null mice.
2) D2 activity is normal in the cortex, cerebellum, and hippocampus, and total brain RC3-neurogranin messenger RNA levels are not altered.
3) T4 levels measured in the cortex, cerebellum, and hippocampus are normal. However T4 and T3 levels in the choroid plexus are only 14% and 48% of the normal values, respectively.
4) T3 levels are normal in the brain parenchyma.
The data presented here suggest that TTR influences thyroid hormone levels in the choroid plexus, but not in the brain. Interference with the blood-choroid-plexus-CSF-TTR-mediated route of T4 entry into the brain caused by the absence of TTR does not produce measurable features of hypothyroidism. It thus appears that TTR is not required for T4 entry or for maintenance of the euthyroid state in the mouse brain.
Identification of monocarboxylate transporter 8 as a specific thyroid hormone transporter

E.C.H. Friesema, S Ganguly, A. Abdalla, J.E.M. Fox, AP. Halestrap, and TJ. Visser
J Biol Chem 2003; Manuscript M300909200
http://dx.doi.org/10.1074/jbc.M300909200

Transport of thyroid hormone across the cell membrane is required for its action and

metabolism. Recently, a T-type amino acid transporter was cloned which transports aromatic amino acids but not iodothyronines. This transporter belongs to the monocarboxylate transporter (MCT) family, and is most homologous with MCT8 (SLC16A2). Therefore, we cloned rat MCT8, and tested it for thyroid hormone transport in Xenopus laevis oocytes. Oocytes were injected with rat MCT8 cRNA, and after 3 days immunofluorescence microscopy demonstrated expression of the protein at the plasma membrane. MCT8 cRNA induced a ~10-fold increase in uptake of 10 nM 125I-labeled thyroxine (T4), 3,3′,5-triiodothyronine (T3), 3,3′,5′-triiodothyronine (rT3) and 3,3′-diiodothyronine. Due to the rapid uptake of the ligands, transport was only linear with time for <4 min. MCT8 did not transport Leu, Phe, Trp or Tyr. [125I]T4 transport was strongly inhibited by L-T4, D-T4, L-T3, D-T3, 3,3’,5-triiodothyroacetic acid, N-bromoacetyl-T3, and bromosulfophthalein. T3 transport was less affected by these inhibitors. Iodothyronine uptake in uninjected oocytes was reduced by albumin but the stimulation induced by MCT8 was markedly increased. Saturation analysis provided apparent Km values of 2-5 μM for T4, T3 and rT3. Immunohistochemistry showed high expression in liver, kidney, brain and heart. In conclusion, we have identified MCT8 as a very active and specific thyroid hormone transporter.
Thyroid hormones,T3 andT4, in the brain
Amy C. Schroeder and Martin L. Privalsky
Front Endocr Mar 2014; 5 article 40.  http://dx.doi.org:/10.3389/fendo.2014.00040

Thyroid hormones (THs) are essential for fetal and post-natal nervous system development and also play an important role in the maintenance of adult brain function. Of the two major THs, T4 (3,5,30,50-tetraiodo-l-thyronine) is classically viewed as an pro-hormone that must be converted toT3 (3,5,30-tri-iodo-l-thyronine) via tissue-level deiodinases for biological activity. THs primarily mediate their effects by binding to thyroid hormone receptor (TR) isoforms, predominantly TRα1 and TRβ1, which are expressed in different tissues and exhibit distinctive roles in endocrinology. Notably, the ability to respond toT4 and toT3 differs for the two TR isoforms, with TRα1 generally more responsive to T4 than TRβ1. TRα1 is also the most abundantly expressed TR isoform in the brain, encompassing 70–80% of all TR expression in this tissue. Conversion of T4 into T3 via deiodinase 2 in astrocytes has been classically viewed as critical for generating local T3 for neurons. However, deiodinase-deficient mice do not exhibit obvious defectives in brain development or function. Considering that TRα1 is well-established as the predominant isoform in brain, and that TRα1 responds to both T3 and T4, we suggest T4 may play a more active role in brain physiology than has been previously accepted.
Thyroid hormone action: astrocyte–neuron communication

Beatriz Morte and Juan Bernal
Front Endocr May 2014; 5, Article 82 http://dx.doi.org:/10.3389/fendo.2014.00082

Thyroid hormone (TH) action is exerted mainly through regulation of gene expression by binding of T3 to the nuclear receptors.T4 plays an important role as a source of intracellular T3 in the central nervous system via the action of the type 2 deiodinase (D2), expressed in the astrocytes. A model of T3 availability to neural cells has been proposed and validated. The model contemplates that brain T3 has a double origin: a fraction is available directly from the circulation, and another is produced locally from T4 in the astrocytes by D2. The fetal brain depends almost entirely on theT3 generated locally. The contribution of systemic T3 increases subsequently during development to account for approximately 50% of total brain T3 in the late postnatal and adult stages. In this article, we review the experimental data in support of this model, and how the factors affectingT3 availability in the brain, such as deiodinases and transporters, play a decisive role in modulating local TH action during development.
The Significance of Thyroid Hormone Transporters in the Brain

Juan Bernal
Endocr Apr 2005; 146(4):1698–1700. http://dx.doi.org:/10.1210/en.2005-0134

The MCT family comprises up to 14 members, some of which are involved in the transport of important substrates for the brain such as lactate and pyruvate. MCT8 has been shown to act as a specific transporter for T4 and T3 and displays slightly higher affinity for T3. Heuer et al. have also studied the regional expression of MCT8 mRNA. In addition to high expression levels in the choroid plexus, they found that MCT8 is expressed in neurons of the neocortex, hippocampus, basal ganglia, amygdala, hypothalamus, and the Purkinje cells of the cerebellum, all regions known to be sensitive to thyroid hormones. Expression of MCT8 in neurons suggests that neuronal uptake of the T3 produced in astrocytes is facilitated by this transporter.
The physiological significance ofMCT8 as a transporter for thyroid hormone is supported by the finding of mutations in humans by Dumitrescu et al. and Friesema et al.  The syndrome affects children from an early age and consists of severe developmental delay and neurological damage together with an unusually altered pattern of thyroid hormone levels in blood. The patients presented low total and free T4, high total and free T3, and low rT3. TSH was moderately elevated in two of the patients and normal or slightly elevated in the other five. Inactivating mutations of the MCT8 transporter could result in the altered thyroid hormone levels. In vitro uptake of T4 and T3 by fibroblasts isolated from affected males was strongly reduced, and intracellular D2 was increased 6- to 8-fold. It is thus hypothesized that the resulting increase in intracellularly generated T3 accumulates in blood because of its poor reuptake into cells.
The second trimester is also the period when thyroid hormone receptors increase in concentration in the brain. If MCT8 is needed at this stage of development for T3 entry into neurons, mutations of the transporter could interfere with T3-dependent developmental processes. Knowledge of the ontogenetic patterns of MCT8 in the human fetal brain would certainly be helpful. On the other hand, there is also the possibility that MCT8 mutations interfere with transport of other substrates for brain metabolism that could be even more important than T3 in determining the severity and outcome of the syndrome. Other members of the family transport metabolic substrates such as pyruvate and lactate, but MCT8 so far appears to be specific for iodothyronines

Peripheral markers of thyroid function: The effect of T4 monotherapy versus T4/T3 combination therapy in hypothyroid subjects: A randomized cross-over study

Ulla Schmidt, B Nygaard, EW Jensen, J Kvetny, A Jarløv, and Jens Faber
Endocrine Connections Jan 10, 2013 http://dx.doi.org:/10.1530/EC-12-0

Background: A recent randomized controlled trial suggests that hypothyroid subjects may find L-T4 and L-T3 combination therapy to be

superior to L-T4 monotherapy in terms of quality of life, suggesting that the brain registered increased T3 availability during the

combination therapy.

Hypothesis: Peripheral tissue might also be stimulated during T4/T3 combination therapy compared to T4 monotherapy.
Methods: Serum levels of Sex Hormone-Binding Globulin (SHBG), pro-collagen-1-N-terminal peptide (PINP), and N-terminal pro-brain natriuretic peptide (NT-proBNP) (representing hepatocyte, osteoblast, and cardiomyocyte stimulation, respectively) were measured in 26 hypothyroid subjects in a double blind, randomized, cross-over trial, which compared the replacement therapy with T4/T3 in combination (50 Fg T4 was substituted with 20 Fg T3) to T4 alone (once daily regimens). This was performed to obtain unaltered serum thyroid stimulating hormone (TSH) levels during the trial and between the two treatment groups. Blood sampling was performed 24 hours after the last intake of thyroid hormone medication.
Results: TSH remained unaltered between the groups ((median) 0.83 vs. 1.18 mU/l in T4/T3 combination and T4 mono-therapy, respectively; p=0.534). SHBG increased from (median) 75 nmol/l at baseline to 83 nmol/l in the T4/T3 group (p=0.015), but remained unaltered in the T4 group (67 nmol/l); thus, it was higher in the T4/T3 vs. T4 group (p=0.041). PINP levels were higher in the T4/T3 therapy (48 vs. 40 Fg/l (p<0.001)). NT-proBNP did not differ between the groups. Conclusions: T4/T3 combination therapy in hypothyroidism seems to have more metabolic effects than the T4 monotherapy.
Stimulatory effects of thyroid hormone on brain angiogenesis in vivo and in vitro

Liqun Zhang, CM Cooper-Kuhn, U Nannmark, K Blomgren and HG Kuhn
J Cereb Blood Flow & Metab 2010; 30:323–335. http://dx.doi.org:/10.1038/jcbfm.2009.216

Thyroid hormone is critical for the proper development of the central nervous system. However, the specific role of thyroid hormone on brain angiogenesis remains poorly understood. Treatment of rats from birth to postnatal day 21 (P21) with propylthiouracil (PTU), a reversible blocker of triiodothyronine (T3) synthesis, resulted in decreased brain angiogenesis, as indicated by reduced complexity and density of microvessels. However, when PTU was withdrawn at P22, these parameters were fully recovered by P90. These changes were paralleled by an  altered expression of vascular endothelial growth factor A (Vegfa) and basic fibroblast growth factor (Fgf2). Physiologic concentrations of T3 and thyroxine (T4) stimulated proliferation and tubulogenesis of rat brain derived endothelial (RBE4) cells in vitro. Protein and mRNA levels of VEGF-A and FGF-2 increased after T3 stimulation of RBE4 cells. The thyroid hormone receptor blocker NH-3 abolished T3-induced Fgf2 and Vegfα upregulation, indicating a receptor-mediated effect. Thyroid hormone inhibited the apoptosis in RBE4 cells and altered mRNA levels of apoptosis-related genes, namely Bcl2 and Bad. The present results show that thyroid hormone has a substantial impact on vasculature development in the brain. Pathologically altered vascularization could, therefore, be a contributing factor to the neurologic deficits induced by thyroid hormone deficiency.

Molecules important for thyroid hormone

synthesis and action – known facts and future perspectives

Klaudia Brix, Dagmar Führer, Heike Biebermann
Thyroid Research 2011, 4(Suppl 1):S9 http://www.thyroidresearchjournal.com/content/4/S1/S9

Thyroid hormones are of crucial importance for the functioning of nearly every organ. Remarkably, disturbances of thyroid hormone synthesis and function are among the most common endocrine disorders affecting approximately one third of the working German population. Over the last ten years our understanding of biosynthesis and functioning of these hormones has increased tremendously. This includes the identification of proteins involved in thyroid hormone biosynthesis like Thox2 and Dehal where mutations in these genes are responsible for certain degrees of hypothyroidism. One of the most important findings was the identification of a specific transporter for triiodothyronine (T3), the monocarboxylate transporter 8 (MCT8) responsible for directed transport of T3 into target cells and for export of thyroid hormones out of thyroid epithelial cells. Genetic disturbances of MCT8 in patients result in a biochemical constellation of high T3 levels in combination with low or normal TSH and thyroxine levels leading to a new syndrome of severe X-linked mental retardation. Importantly mice lacking MCT8 presented only with a mild phenotype, indicating that compensatory mechanisms exist in mice. Moreover, it has become clear that not only genomic actions of T3 exist. T3 is also capable to activate adhesion receptors and it signals via activation of PI3K and MAPK pathways. Most recently, thyroid hormone derivatives were identified, the thyronamines which are decarboxylated thyroid hormones initiating physiological actions like lowering body temperature and heart rate, thereby acting in opposite direction to the classical thyroid hormones. So far it is believed that thyronamines function via the activation of a G-protein coupled receptor, TAAR1. The objective of this review is to summarize the recent findings in thyroid hormone synthesis and action and to discuss their implications for diagnosis of thyroid disease and for treatment of patients.

Retinoic Acid Induces Expression of the Thyroid Hormone Transporter, Monocarboxylate Transporter 8 (Mct8)

T Kogai, Yan-Yun Liu, LL Richter, K Mody, H Kagechika, and GA Brent
J Biol Chem Jun 2010. Manuscript M110.123158
http://www.jbc.org/cgi/doi/10.1074/jbc.M110.123158

Retinoic acid (RA) and thyroid hormone are critical for differentiation and organogenesis in the embryo. The monocarboxylate transporter-8 (Mct8), expressed predominantly in brain and placenta, mediates thyroid hormone uptake from the circulation and is required for normal neural development. RA induces differentiation of F9 mouse teratocarcinoma cells towards neurons as well as extraembryonal endoderm. We hypothesized that Mct8 is functionally expressed in F9 cells and induced by RA.  All trans RA (tRA), and other RA receptor (RAR) agonists, dramatically (> 300-fold) induced Mct8. tRA treatment significantly increased uptake of triiodothyronine and thyroxine (4.1 fold and 4.3 fold, respectively), which was abolished by a selective Mct8 inhibitor, bromosulfophthalein. Sequence inspection of the Mct8 promoter region and
5′-rapid amplification of cDNA ends (5’-RACE) PCR analysis in F9 cells identified
11 transcription start sites and a proximal Sp1 site, but no TATA-box.  tRA significantly enhanced Mct8 promoter activity through a consensus RA responsive element located 6.6 kilobases upstream of the coding region. Chromatin immunoprecipitation assay demonstrated binding of RAR and retinoid-X receptor (RXR) to the RA response element. The promotion of thyroid hormone uptake through the transcriptional up-regulation of Mct8 by RAR is likely to be important for extraembryonic endoderm development and neural differentiation. This finding demonstrates crosstalk between RA signaling and thyroid hormone signaling in early development at the level of the thyroid hormone transporter.
Abnormal thyroid hormone metabolism in mice lacking the monocarboxylate transporter 8

Marija Trajkovic, Theo J. Visser, Jens Mittag, Sigrun Horn, et al.
J. Clin. Invest.  2007; 117:627–635. http://dx.doi.org:/10.1172/JCI28253

In humans, inactivating mutations in the gene of the thyroid hormone transporter monocarboxylate transporter 8 (MCT8; SLC16A2) lead to severe forms of psychomotor retardation combined with imbalanced thyroid hormone serum levels. The MCT8-null mice described here, however, developed without overt deficits but also exhibited distorted 3,5,3′-triiodothyronine (T3) and thyroxine (T4) serum levels, resulting in increased hepatic activity of type 1 deiodinase (D1). In the mutants’ brains, entry of T4 was not affected, but uptake of T3 was diminished. Moreover, the T4 and T3 content in the brain of MCT8-null mice was decreased, the activity of D2 was increased, and D3 activity was decreased, indicating the hypothyroid state of this tissue. In the CNS, analysis of T3 target genes revealed that in the mutants, the neuronal T3 uptake was impaired in an area-specific manner, with strongly elevated thyrotropin-releasing hormone transcript levels in the hypothalamic paraventricular nucleus and slightly decreased RC3 mRNA expression in striatal neurons; however, cerebellar Purkinje cells appeared unaffected, since they did not exhibit dendritic outgrowth defects and responded normally to T3 treatment in vitro.
In conclusion, the circulating thyroid hormone levels of MCT8-null mice closely resemble those of humans with MCT8 mutations, yet in the mice, CNS development is only partially affected.
3-Monoiodothyronamine: the rationale for its action as an endogenous adrenergic-blocking neuromodulator

HS Gompf, JH Greenberg, G Aston-Jones, A Ianculescu, TS Scanlan, and MB Dratman
Brain Res. 2010 Sep 10; 1351: 130–140. http://dx.doi.org:/10.1016/j.brainres.2010.06.067

The investigations reported here were designed to gain insights into the role of
3-monoiodothyronamine (T1AM) in the brain, where the amine was originally identified and characterized.
Extensive deiodinase studies indicated that T1AM was derived from the T4 metabolite, reverse triiodothyronine (revT3), while functional studies provided well-confirmed evidence that T1AM has strong adrenergic blocking effects. Because a state of adrenergic overactivity prevails when triiodothyronine (T3) concentrations becomes excessive, the possibility that T3’s metabolic partner, revT3, might give rise to an antagonist of those T3 actions was thought to be reasonable.
All T1AM studies thus far have required use of pharmacological doses.
Therefore we considered that choosing a physiological site of action was a priority and focused on the locus coeruleus (LC), the major noradrenergic control center in the brain. Site-directed injections of T1AM into the LC elicited a significant, dose-dependent neuronal firing rate change in a subset of adrenergic neurons with an EC50=2.7 μM, a dose well within the physiological range. Further evidence for its physiological actions came from autoradiographic images obtained following intravenous carrier-free 125I-labeled T1AM injection. These showed that the amine bound with high affinity to the LC and to other selected brain nuclei, each of which is both an LC target and a known T3 binding site. This new evidence points to a physiological role for T1AM as an endogenous adrenergic-blocking neuromodulator in the central noradrenergic system.

Thyroid hormones are transported through the blood-brain barrier

Thyroid hormones are transported through the blood-brain barrier

Thyroid hormones are transported through the blood-brain barrier (OATP) or the blood-CSF barrier (OATP and MCT8). In the astrocytes and tanycytes T4 is converted to T3 which then enters the neurons through MCT8. In the neurons both T4 and T3 are degraded by D3. T3 from the tanycytes may reach the portal vessels in the median eminence. Other transporters may be present on the astrocyte or tanycyte membranes. In most cases the transport could be bidirectional, although only one direction is shown.
Juan Bernal – Instituto de Investigaciones Biomedicas – 28029 Madrid, Spain

the interactions of maternal, placental and fetal thyroid

the interactions of maternal, placental and fetal thyroid

Old and new concepts of thyroid hormone action.

A: Old concept of thyroid hormone action. In former times it was assumed that thyroid hormones are able to pass the plasma membrane by passive transport. Once in the cytosol T4 is deiodinated to T3 which exerts genomic effects by binding to the thyroid hormone receptor (TR). After hetero-dimerization with other nuclear receptors like retinoic X receptor (RXR), transcriptional regulation is initiated resulting in activation or inactivation of target genes.
B: New concepts of thyroid hormone action. Thyroid hormones enter a target cell via specific transporters, e.g. T3 uses the monocarboxylate transporter MCT8 while T4 entry is mediated by Lat2 or Oatp14. Moreover, T3 can interact with avb3 integrins to induce ERK1/2 signalling. Cytosolic T3 exerts genomic effects but can additionally also act by non-genomic means after TR binding and activation of down-stream PI-3 kinase. Likewise, the naturally occurring iodothyronine T2 is believed to stimulate metabolic rates via mitochondrial pathways, thereby bypassing genomic regulation. Besides thyroid hormones, derivatives like the thyronamines T1AM or T0AM, modulate the action of T3, e.g. counter-acting its effects in certain target cells. Thyronamines (TAMs) bind to and activate G-protein coupled receptors (GPCRs) of the trace amine associated receptor (TAAR) family. So far, it is only known that TAAR1 is activated by TAMs and signals via adenylylcyclase (AC) activation with subsequent rise of cAMP levels. However other GPCRs are likely targets for thyroid hormone derivatives

Brix et al.: Molecules important for thyroid hormone synthesis and action – known facts and future perspectives. Thyroid Research 2011 4(Suppl 1):S9.
http://dx.doi.org:/10.1186/1756-6614-4-S1-S9

Read Full Post »

%d bloggers like this: