Feeds:
Posts
Comments

Archive for August, 2014

Compilation of References in Leaders in Pharmaceutical Intelligence about proteomics, metabolomics, signaling pathways, and cell regulation

Compilation of References in Leaders in Pharmaceutical Intelligence about
proteomics, metabolomics, signaling pathways, and cell regulation

Curator: Larry H. Bernstein, MD, FCAP

 

Proteomics

  1. The Human Proteome Map Completed
    Reporter and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/28/the-human-proteome-map-completed/
  1. Proteomics – The Pathway to Understanding and Decision-making in Medicine
    Author and Curator, Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/06/24/proteomics-the-pathway-to-understanding-and-decision-making-in-medicine/
  1. Advances in Separations Technology for the “OMICs” and Clarification of Therapeutic Targets
    Author and Curator, Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2012/10/22/advances-in-separations-technology-for-the-omics-and-clarification-of-therapeutic-targets/
  1. Expanding the Genetic Alphabet and Linking the Genome to the Metabolome
    Author and Curator, Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2012/09/24/expanding-the-genetic-alphabet-and-linking-the-genome-to-the-metabolome/
  1. Synthesizing Synthetic Biology: PLOS Collections
    Reporter: Aviva Lev-Ari
    http://pharmaceuticalintelligence.com/2012/08/17/synthesizing-synthetic-biology-plos-collections/

 

Metabolomics

  1. Extracellular evaluation of intracellular flux in yeast cells
    Larry H. Bernstein, MD, FCAP, Reviewer and Curator
    http://pharmaceuticalintelligence.com/2014/08/25/extracellular-evaluation-of-intracellular-flux-in-yeast-cells/ 
  2. Metabolomic analysis of two leukemia cell lines. I.
    Larry H. Bernstein, MD, FCAP, Reviewer and Curator
    http://pharmaceuticalintelligence.com/2014/08/23/metabolomic-analysis-of-two-leukemia-cell-lines-_i/ 
  3. Metabolomic analysis of two leukemia cell lines. II.
    Larry H. Bernstein, MD, FCAP, Reviewer and Curator
    http://pharmaceuticalintelligence.com/2014/08/24/metabolomic-analysis-of-two-leukemia-cell-lines-ii/ 
  4. Metabolomics, Metabonomics and Functional Nutrition: the next step in nutritional metabolism and biotherapeutics
    Reviewer and Curator, Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/22/metabolomics-metabonomics-and-functional-nutrition-the-next-step-in-nutritional-metabolism-and-biotherapeutics/ 
  5. Buffering of genetic modules involved in tricarboxylic acid cycle metabolism provides homeomeostatic regulation
    Larry H. Bernstein, MD, FCAP, Reviewer and curator
    http://pharmaceuticalintelligence.com/2014/08/27/buffering-of-genetic-modules-involved-in-tricarboxylic-acid-cycle-metabolism-provides-homeomeostatic-regulation/

 

Metabolic Pathways

  1. Pentose Shunt, Electron Transfer, Galactose, more Lipids in brief
    Reviewer and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/21/pentose-shunt-electron-transfer-galactose-more-lipids-in-brief/
  2. Mitochondria: More than just the “powerhouse of the cell”
    Reviewer and Curator: Ritu Saxena
    http://pharmaceuticalintelligence.com/2012/07/09/mitochondria-more-than-just-the-powerhouse-of-the-cell/
  3. Mitochondrial fission and fusion: potential therapeutic targets?
    Reviewer and Curator: Ritu saxena
    http://pharmaceuticalintelligence.com/2012/10/31/mitochondrial-fission-and-fusion-potential-therapeutic-target/ 
  4. Mitochondrial mutation analysis might be “1-step” away
    Reviewer and Curator: Ritu Saxena
    http://pharmaceuticalintelligence.com/2012/08/14/mitochondrial-mutation-analysis-might-be-1-step-away/
  5. Selected References to Signaling and Metabolic Pathways in PharmaceuticalIntelligence.com
    Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/14/selected-references-to-signaling-and-metabolic-pathways-in-leaders-in-pharmaceutical-intelligence/
  6. Metabolic drivers in aggressive brain tumors
    Prabodh Kandal, PhD
    http://pharmaceuticalintelligence.com/2012/11/11/metabolic-drivers-in-aggressive-brain-tumors/ 
  7. Metabolite Identification Combining Genetic and Metabolic Information: Genetic association links unknown metabolites to functionally related genes
    Author and Curator: Aviva Lev-Ari, PhD, RD
    http://pharmaceuticalintelligence.com/2012/10/22/metabolite-identification-combining-genetic-and-metabolic-information-genetic-association-links-unknown-metabolites-to-functionally-related-genes/
  8. Mitochondria: Origin from oxygen free environment, role in aerobic glycolysis, metabolic adaptation
    Author and curator:Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2012/09/26/mitochondria-origin-from-oxygen-free-environment-role-in-aerobic-glycolysis-metabolic-adaptation/
  9. Therapeutic Targets for Diabetes and Related Metabolic Disorders
    Reporter, Aviva Lev-Ari, PhD, RD
    http://pharmaceuticalintelligence.com/2012/08/20/therapeutic-targets-for-diabetes-and-related-metabolic-disorders/
  10. Buffering of genetic modules involved in tricarboxylic acid cycle metabolism provides homeomeostatic regulation
    Larry H. Bernstein, MD, FCAP, Reviewer and curator
    http://pharmaceuticalintelligence.com/2014/08/27/buffering-of-genetic-modules-involved-in-tricarboxylic-acid-cycle-metabolism-provides-homeomeostatic-regulation/
  11. The multi-step transfer of phosphate bond and hydrogen exchange energy
    Curator:Larry H. Bernstein, MD, FCAP,
    http://pharmaceuticalintelligence.com/2014/08/19/the-multi-step-transfer-of-phosphate-bond-and-hydrogen-exchange-energy/
  12. Studies of Respiration Lead to Acetyl CoA
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/18/studies-of-respiration-lead-to-acetyl-coa/
  13. Lipid Metabolism
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/15/lipid-metabolism/
  14. Carbohydrate Metabolism
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/13/carbohydrate-metabolism/
  15. Prologue to Cancer – e-book Volume One – Where are we in this journey?
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/04/13/prologue-to-cancer-ebook-4-where-are-we-in-this-journey/
  16. Introduction – The Evolution of Cancer Therapy and Cancer Research: How We Got Here?
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/04/04/introduction-the-evolution-of-cancer-therapy-and-cancer-research-how-we-got-here/
  17. Inhibition of the Cardiomyocyte-Specific Kinase TNNI3K
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2013/11/01/inhibition-of-the-cardiomyocyte-specific-kinase-tnni3k/
  18. The Binding of Oligonucleotides in DNA and 3-D Lattice Structures
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2013/05/15/the-binding-of-oligonucleotides-in-dna-and-3-d-lattice-structures/
  19. Mitochondrial Metabolism and Cardiac Function
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2013/04/14/mitochondrial-metabolism-and-cardiac-function/
  20. How Methionine Imbalance with Sulfur-Insufficiency Leads to Hyperhomocysteinemia
    Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2013/04/04/sulfur-deficiency-leads_to_hyperhomocysteinemia/
  21. AMPK Is a Negative Regulator of the Warburg Effect and Suppresses Tumor Growth In Vivo
    Author and Curator: SJ. Williams
    http://pharmaceuticalintelligence.com/2013/03/12/ampk-is-a-negative-regulator-of-the-warburg-effect-and-suppresses-tumor-growth-in-vivo/
  22. A Second Look at the Transthyretin Nutrition Inflammatory Conundrum
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2012/12/03/a-second-look-at-the-transthyretin-nutrition-inflammatory-conundrum/
  23. Overview of Posttranslational Modification (PTM)
    Writer and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/07/29/overview-of-posttranslational-modification-ptm/
  24. Malnutrition in India, high newborn death rate and stunting of children age under five years
    Writer and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/07/15/malnutrition-in-india-high-newborn-death-rate-and-stunting-of-children-age-under-five-years/
  25. Update on mitochondrial function, respiration, and associated disorders
    Writer and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/07/08/update-on-mitochondrial-function-respiration-and-associated-disorders/
  26. Omega-3 fatty acids, depleting the source, and protein insufficiency in renal disease
    Larry H. Bernstein, MD, FCAP, Curator
    http://pharmaceuticalintelligence.com/2014/07/06/omega-3-fatty-acids-depleting-the-source-and-protein-insufficiency-in-renal-disease/ 
  27. Late Onset of Alzheimer’s Disease and One-carbon Metabolism
    Reporter and Curator: Dr. Sudipta Saha, Ph.D.
    http://pharmaceuticalintelligence.com/2013/05/06/alzheimers-disease-and-one-carbon-metabolism/
  28. Problems of vegetarianism
    Reporter and Curator: Dr. Sudipta Saha, Ph.D.
    http://pharmaceuticalintelligence.com/2013/04/22/problems-of-vegetarianism/

 

Signaling Pathways

  1. Introduction to e-Series A: Cardiovascular Diseases, Volume Four Part 2: Regenerative Medicine
    Larry H. Bernstein, MD, FCAP, writer, and Aviva Lev- Ari, PhD, RN  http://pharmaceuticalintelligence.com/2014/04/27/larryhbernintroduction_to_cardiovascular_diseases-translational_medicine-part_2/
  2. Epilogue: Envisioning New Insights in Cancer Translational Biology
    Series C: e-Books on Cancer & Oncology
    Author & Curator: Larry H. Bernstein, MD, FCAP, Series C Content Consultant
    http://pharmaceuticalintelligence.com/2014/03/29/epilogue-envisioning-new-insights/
  3. Ca2+-Stimulated Exocytosis:  The Role of Calmodulin and Protein Kinase C in Ca2+ Regulation of Hormone and Neurotransmitter  Writer and Curator: Larry H Bernstein, MD, FCAP and Curator and Content Editor: Aviva Lev-Ari, PhD, RN
    http://pharmaceuticalintelligence.com/2013/12/23/calmodulin-and-protein-kinase-c-drive-the-ca2-regulation-of-hormone-and-neurotransmitter-release-that-triggers-ca2-stimulated-exocy
  4. Cardiac Contractility & Myocardial Performance: Therapeutic Implications of Ryanopathy (Calcium Release-related Contractile Dysfunction) and Catecholamine Responses
    Author, and Content Consultant to e-SERIES A: Cardiovascular Diseases: Justin Pearlman, MD, PhD, FACC
    Author and Curator: Larry H Bernstein, MD, FCAP and Article Curator: Aviva Lev-Ari, PhD, RN
    http://pharmaceuticalintelligence.com/2013/08/28/cardiac-contractility-myocardium-performance-ventricular-arrhythmias-and-non-ischemic-heart-failure-therapeutic-implications-for-cardiomyocyte-ryanopathy-calcium-release-related-contractile/
  5. Role of Calcium, the Actin Skeleton, and Lipid Structures in Signaling and Cell Motility
    Author and Curator: Larry H Bernstein, MD, FCAP Author: Stephen Williams, PhD, and Curator: Aviva Lev-Ari, PhD, RN
    http://pharmaceuticalintelligence.com/2013/08/26/role-of-calcium-the-actin-skeleton-and-lipid-structures-in-signaling-and-cell-motility/
  6. Identification of Biomarkers that are Related to the Actin Cytoskeleton
    Larry H Bernstein, MD, FCAP, Author and Curator
    http://pharmaceuticalintelligence.com/2012/12/10/identification-of-biomarkers-that-are-related-to-the-actin-cytoskeleton/
  7. Advanced Topics in Sepsis and the Cardiovascular System at its End Stage
    Author and Curator: Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2013/08/18/advanced-topics-in-Sepsis-and-the-Cardiovascular-System-at-its-End-Stage/
  8. The Delicate Connection: IDO (Indolamine 2, 3 dehydrogenase) and Cancer Immunology
    Demet Sag, PhD, Author and Curator
    http://pharmaceuticalintelligence.com/2013/08/04/the-delicate-connection-ido-indolamine-2-3-dehydrogenase-and-immunology/
  9. IDO for Commitment of a Life Time: The Origins and Mechanisms of IDO, indolamine 2, 3-dioxygenase
    Demet Sag, PhD, Author and Curator
    http://pharmaceuticalintelligence.com/2013/08/04/ido-for-commitment-of-a-life-time-the-origins-and-mechanisms-of-ido-indolamine-2-3-dioxygenase/
  10. Confined Indolamine 2, 3 dioxygenase (IDO) Controls the Homeostasis of Immune Responses for Good and Bad
    Author and Curator: Demet Sag, PhD, CRA, GCP
    http://pharmaceuticalintelligence.com/2013/07/31/confined-indolamine-2-3-dehydrogenase-controls-the-hemostasis-of-immune-responses-for-good-and-bad/
  11. Signaling Pathway that Makes Young Neurons Connect was discovered @ Scripps Research Institute
    Reporter: Aviva Lev-Ari, PhD, RN
    http://pharmaceuticalintelligence.com/2013/06/26/signaling-pathway-that-makes-young-neurons-connect-was-discovered-scripps-research-institute/
  12. Naked Mole Rats Cancer-Free
    Writer and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2013/06/20/naked-mole-rats-cancer-free/
  13. Amyloidosis with Cardiomyopathy
    Writer and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2013/03/31/amyloidosis-with-cardiomyopathy/
  14. Liver endoplasmic reticulum stress and hepatosteatosis
    Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2013/03/10/liver-endoplasmic-reticulum-stress-and-hepatosteatosis/
  15. The Molecular Biology of Renal Disorders: Nitric Oxide – Part III
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2012/11/26/the-molecular-biology-of-renal-disorders/
  16. Nitric Oxide Function in Coagulation – Part II
    Curator and Author: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2012/11/26/nitric-oxide-function-in-coagulation/
  17. Nitric Oxide, Platelets, Endothelium and Hemostasis
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2012/11/08/nitric-oxide-platelets-endothelium-and-hemostasis/
  18. Interaction of Nitric Oxide and Prostacyclin in Vascular Endothelium
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2012/09/14/interaction-of-nitric-oxide-and-prostacyclin-in-vascular-endothelium/
  19. Nitric Oxide and Immune Responses: Part 1
    Curator and Author:  Aviral Vatsa PhD, MBBS
    http://pharmaceuticalintelligence.com/2012/10/18/nitric-oxide-and-immune-responses-part-1/
  20. Nitric Oxide and Immune Responses: Part 2
    Curator and Author:  Aviral Vatsa PhD, MBBS
    http://pharmaceuticalintelligence.com/2012/10/28/nitric-oxide-and-immune-responses-part-2/
  21. Nitric Oxide and iNOS have Key Roles in Kidney Diseases – Part II
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2012/11/26/nitric-oxide-and-inos-have-key-roles-in-kidney-diseases/
  22. New Insights on Nitric Oxide donors – Part IV
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2012/11/26/new-insights-on-no-donors/
  23. Crucial role of Nitric Oxide in Cancer
    Curator and Author: Ritu Saxena, Ph.D.
    http://pharmaceuticalintelligence.com/2012/10/16/crucial-role-of-nitric-oxide-in-cancer/
  24. Nitric Oxide has a ubiquitous role in the regulation of glycolysis -with a concomitant influence on mitochondrial function
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2012/09/16/nitric-oxide-has-a-ubiquitous-role-in-the-regulation-of-glycolysis-with-a-concomitant-influence-on-mitochondrial-function/
  25. Nitric Oxide and Immune Responses: Part 2
    Author and Curator: Aviral Vatsa, PhD, MBBS
    http://pharmaceuticalintelligence.com/2012/10/28/nitric-oxide-and-immune-responses-part-2/
  26. Mitochondrial Damage and Repair under Oxidative Stress
    Author and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2012/10/28/mitochondrial-damage-and-repair-under-oxidative-stress/
  27. Is the Warburg Effect the cause or the effect of cancer: A 21st Century View?
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2012/10/17/is-the-warburg-effect-the-cause-or-the-effect-of-cancer-a-21st-century-view/
  28. Targeting Mitochondrial-bound Hexokinase for Cancer Therapy
    Curator and Author: Ziv Raviv, PhD, RN 04/06/2013
    http://pharmaceuticalintelligence.com/2013/04/06/targeting-mitochondrial-bound-hexokinase-for-cancer-therapy/
  29. Ubiquinin-Proteosome pathway, autophagy, the mitochondrion, proteolysis and cell apoptosis
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2012/10/30/ubiquinin-proteosome-pathway-autophagy-the-mitochondrion-proteolysis-and-cell-apoptosis/
  30. Ubiquitin-Proteosome pathway, Autophagy, the Mitochondrion, Proteolysis and Cell Apoptosis: Part III
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2013/02/14/ubiquinin-proteosome-pathway-autophagy-the-mitochondrion-proteolysis-and-cell-apoptosis-reconsidered/
  31. Biochemistry of the Coagulation Cascade and Platelet Aggregation – Part I
    Curator and Author: Larry H Bernstein, MD, FACP
    http://pharmaceuticalintelligence.com/2012/11/26/biochemistry-of-the-coagulation-cascade-and-platelet-aggregation/

 

Genomics, Transcriptomics, and Epigenetics

  1. What is the meaning of so many RNAs?
    Writer and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/06/what-is-the-meaning-of-so-many-rnas/
  2. RNA and the transcription the genetic code
    Larry H. Bernstein, MD, FCAP, Writer and Curator
    http://pharmaceuticalintelligence.com/2014/08/02/rna-and-the-transcription-of-the-genetic-code/
  3. A Primer on DNA and DNA Replication
    Writer and Curator: Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/07/29/a_primer_on_dna_and_dna_replication/
  4. Pathology Emergence in the 21st Century
    Author and Curator: Larry Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/03/pathology-emergence-in-the-21st-century/
  5. RNA and the transcription the genetic code
    Writer and Curator, Larry H. Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/02/rna-and-the-transcription-of-the-genetic-code/
  6. Commentary on Biomarkers for Genetics and Genomics of Cardiovascular Disease: Views by Larry H Bernstein, MD, FCAP
    Author: Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/07/16/commentary-on-biomarkers-for-genetics-and-genomics-of-cardiovascular-disease-views-by-larry-h-bernstein-md-fcap/
  7. Observations on Finding the Genetic Links in Common Disease: Whole Genomic Sequencing Studies
    Author an Curator: Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2013/05/18/observations-on-finding-the-genetic-links/
  8. Silencing Cancers with Synthetic siRNAs
    Larry H. Bernstein, MD, FCAP, Reviewer and Curator
    http://pharmaceuticalintelligence.com/2013/12/09/silencing-cancers-with-synthetic-sirnas/
  9. Cardiometabolic Syndrome and the Genetics of Hypertension: The Neuroendocrine Transcriptome Control Points
    Reporter: Aviva Lev-Ari, PhD, RN
    http://pharmaceuticalintelligence.com/2013/12/12/cardiometabolic-syndrome-and-the-genetics-of-hypertension-the-neuroendocrine-transcriptome-control-points/
  10. Developments in the Genomics and Proteomics of Type 2 Diabetes Mellitus and Treatment Targets
    Larry H. Bernstein, MD, FCAP, Reviewer and Curator
    http://pharmaceuticalintelligence.com/2013/12/08/developments-in-the-genomics-and-proteomics-of-type-2-diabetes-mellitus-and-treatment-targets/
  11. CT Angiography & TrueVision™ Metabolomics (Genomic Phenotyping) for new Therapeutic Targets to Atherosclerosis
    Reporter: Aviva Lev-Ari, PhD, RN
    http://pharmaceuticalintelligence.com/2013/11/15/ct-angiography-truevision-metabolomics-genomic-phenotyping-for-new-therapeutic-targets-to-atherosclerosis/
  12. CRACKING THE CODE OF HUMAN LIFE: The Birth of BioInformatics & Computational Genomics
    Genomics Curator, Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/08/30/cracking-the-code-of-human-life-the-birth-of-bioinformatics-computational-genomics/
  13. Big Data in Genomic Medicine
    Author and Curator, Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2012/12/17/big-data-in-genomic-medicine/
  14.  From Genomics of Microorganisms to Translational Medicine
    Author and Curator: Demet Sag, PhD
    http://pharmaceuticalintelligence.com/2014/03/20/without-the-past-no-future-but-learn-and-move-genomics-of-microorganisms-to-translational-medicine/
  15.  Summary of Genomics and Medicine: Role in Cardiovascular Diseases
    Author and Curator, Larry H Bernstein, MD, FCAP
    http://pharmaceuticalintelligence.com/2014/01/06/summary-of-genomics-and-medicine-role-in-cardiovascular-diseases/

Read Full Post »

CRACKING THE CODE OF HUMAN LIFE: The Birth of BioInformatics & Computational Genomics

Author and Curator: Larry H Bernstein, MD, FCAP

 

The previous Part II: Cracking the Code of Human Life,

Part II  From Molecular Biology to Translational Medicine:How Far Have We Come, and Where Does It Lead Us? Is broken into a three part series.

Part II A. “CRACKING THE CODE OF HUMAN LIFE: Milestones along the Way” reviews the Human Genome Project and the decade beyond.

Part IIB. “CRACKING THE CODE OF HUMAN LIFE: The Birth of BioInformatics & Computational Genomics” lays the manifold multivariate systems analytical tools that has moved the science forward to a groung that ensures clinical application.

Part IIC. “CRACKING THE CODE OF HUMAN LIFE: Recent Advances in Genomic Analysis and Disease “ extends the discussion to advances in the management of patients as well as providing a roadmap for pharmaceutical drug targeting.

Part III concludes with Ubiquitin, it’s role in Signaling and Regulatory Control.

This article is a continuation of a previous discussion on the role of genomics in discovery of therapeutic targets titled, Directions for Genomics in Personalized Medicine, which focused on: key drivers of cellular proliferation, stepwise mutational changes coinciding with cancer progression, and potential therapeutic targets for reversal of the process. And it is a direct extension of Cracking the Code of Human Life (Part I): “the initiation phase of molecular biology”.

These articles review a web-like connectivity between inter-connected scientific discoveries, as significant findings have led to novel hypotheses and many expectations over the last 75 years. This largely post WWII revolution has driven our understanding of biological and medical processes at an exponential pace owing to successive discoveries of chemical structure,

  • the basic building blocks of DNA  and proteins,
  • of nucleotide and protein-protein interactions,
  • protein folding, allostericity,
  • genomic structure,
  • DNA replication,
  • nuclear polyribosome interaction, and
  • metabolic control.

In addition, the emergence of methods

  • for copying,
  • removal and
  • insertion, and
  1. improvements in structural analysis as well as
  2. developments in applied mathematics have transformed the research framework.

CRACKING THE CODE OF HUMAN LIFE: The Birth of BioInformatics & Computational Genomics Computational Genomics I. Three-Dimensional Folding and Functional Organization Principles of The Drosophila Genome Sexton T, Yaffe E, Kenigeberg E, Bantignies F,…Cavalli G. Institute de Genetique Humaine, Montpelliere GenomiX, and Weissman Institute, France and Israel. Cell 2012; 148(3): 458-472. http://dx.doi.org/10.1016/j.cell.2012.01.010/

Chromosomes are the physical realization of genetic information and thus

  • form the basis for its readout and propagation.

Here we present a high-resolution chromosomal contact map derived from

  • a modified genome-wide chromosome conformation capture approach
  • applied to Drosophila embryonic nuclei.

the entire genome is linearly partitioned into

  • well-demarcated physical domains that
  • overlap extensively with
  • active and repressive epigenetic marks.

Chromosomal contacts are hierarchically organized between domains.

Global modeling of contact density and clustering of domains show

  • that inactive domains are condensed and
  • confined to their chromosomal territories, whereas
  • active domains reach out of the territory to form
  • remote intra- and interchromosomal contacts.

Moreover, we systematically identify specific

  • long-range intrachromosomal contacts between
  • Polycomb-repressed domains.

Together, these observations allow for

  • quantitative prediction of the Drosophila chromosomal contact map,
  • laying the foundation for detailed studies of
  • chromosome structure and function in
  • a genetically tractable system.

Insert pictures

profiles validate the Hi-C Genome wide map

profiles validate the Hi-C Genome wide map

IIC. “Mr. President; The Genome is Fractal !” Eric Lander

(Science Adviser to the President and Director of Broad Institute) et al.
delivered the message on Science Magazine cover (Oct. 9, 2009) and
generated interest in this by the International HoloGenomics Society at
a Sept meeting.

  • First, it may seem to be trivial to rectify the statement in “About cover”
    of Science Magazine by AAAS. The statement “the Hilbert curve is a
    one-dimensional fractal trajectory” needs mathematical clarification.

While the paper itself does not make this statement, the new Editorship
of the AAAS Magazine might be even more advanced if the previous
Editorship did not reject (without review) a Manuscript by 20+ Founders
of (formerly) International PostGenetics Society in December, 2006.

  • Second, it may not be sufficiently clear for the reader that the
    reasonable requirement for the DNA polymerase to crawl along
    a “knot-free” (or “low knot”) structure does not need fractals. A
    “knot-free” structure could be spooled by an ordinary “knitting globule”
    (such that the DNA polymerase does not bump into a “knot” when
    duplicating the strand; just like someone knitting can go through
    the entire thread without encountering an annoying knot): Just to
    be “knot-free” you don’t need fractals.

Note, however, that the “strand” can be accessed only at its beginning –
it is impossible to e.g.

  • to pluck a segment from deep inside the “globulus”.

This is where certain fractals provide a major advantage – that could be

  • the “Eureka” moment for many readers.

For instance, the mentioned Hilbert-curve is not only “knot free” – but

  • provides an easy access to “linearly remote” segments of the strand.

If the Hilbert curve starts from the lower right corner and ends at the lower left corner,

  • for instance the path shows the very easy access of what would be the mid-point
  • if the Hilbert-curve is measured by
  • the Euclidean distance along the zig-zagged path.

Likewise, even the path from the beginning of the Hilbert-curve is about equally easy to access –

  • easier than to reach from the origin a point that is about 2/3 down the path.

The Hilbert-curve provides an easy access between two points

  • within the “spooled thread”;

from a point that is about 1/5 of the overall length

  • to about 3/5 is also in a “close neighborhood”.

This may be the “Eureka-moment” for some readers, to realize that

  • the strand of “the Double Helix” requires quite a finess to fold into
  • the densest possible globuli (the chromosomes) in a clever way
  • that various segments can be easily accessed.

Moreover, in a way that distances

  • between various segments are minimized.

This marvelous fractal structure

  • is illustrated by the 3D rendering of the Hilbert-curve.

Once you observe such fractal structure, you’ll never again think of

  • a chromosome as a “brillo mess”, would you?

It will dawn on you that the genome is orders of magnitudes more

  • finessed than we ever thought so.

Insert picture

profiles validate the Hi-C Genome wide map

profiles validate the Hi-C Genome wide map

Those embarking at a somewhat complex review of some

  • historical aspects of the power of fractals may wish to consult
  • the ouvre of Mandelbrot (also, to celebrate his 85th birthday).

For the more sophisticated readers, even the fairly simple

Hilbert-curve (a representative of the Peano-class) becomes

  • even more stunningly brilliant than just some “see through density”.

Those who are familiar with the classic “Traveling Salesman Problem”

  • know that “the shortest path along which every given n locations can
  • be visited once, and only once” requires fairly sophisticated algorithms
  • (and tremendous amount of computation if n>10 (or much more).

Some readers will be amazed, therefore, that for n=9 the underlying Hilbert-curve

Briefly, the significance of the above realization, that the (recursive)

  1. Fractal Hilbert Curve is intimately connected to the
  2. (recursive) solution of TravelingSalesman Problem,
  3. a core-concept of Artificial Neural Networks summarized below.

Accomplished physicist John Hopfield aroused great excitement in 1982
(already a member of the National Academy of Science)

with his (recursive) design of artificial neural networks and learning algorithms

which were able to find reasonable solutions to combinatorial problems

such as the Traveling SalesmanProblem.
(Book review Clark Jeffries, 1991;  1. J. Anderson, R. Rosenfeld, and
A. Pellionisz (eds.), Neurocomputing 2: Directions for research, MIT
Press, Cambridge, MA, 1990):

“Perceptions were modeled chiefly with neural connections in a

  • “forward” direction: A -> B -* C — D.

The analysis of networks with strong

  • backward coupling proved intractable.

All our interesting results arise as consequences of the strong

  • back-coupling” (Hopfield, 1982).

The Principle of Recursive Genome Function surpassed obsolete

  • axioms that blocked, for half a Century,
  • entry of recursive algorithms to interpretation
  • of the structure-and function of (Holo)Genome.

This breakthrough, by uniting the two largely separate fields of

  • Neural Networks and Genome Informatics,

is particularly important for those who focused on

  • Biological (actually occurring) Neural Networks
  • (rather than abstract algorithms that may not, or
  • because of their core-axioms, simply could not
  • represent neural networks under the governance of DNA information).

IIIA. The FractoGene Decade from Inception in 2002 to Proofs of Concept and
Impending Clinical Applications by 2012

  1. Junk DNA Revisited (SF Gate, 2002)
  2. The Future of Life, 50th Anniversary of DNA (Monterey, 2003)
  3. Mandelbrot and Pellionisz (Stanford, 2004)
  4. Morphogenesis, Physiology and Biophysics (Simons, Pellionisz 2005)
  5. PostGenetics; Genetics beyond Genes (Budapest, 2006)
  6. ENCODE-conclusion (Collins, 2007)
  7. The Principle of Recursive Genome Function (paper, YouTube, 2008)
  8. You Tube Cold Spring Harbor presentation of FractoGene (Cold Spring Harbor, 2009)
  9. Mr. President, the Genome is Fractal! (2009)
  10. HolGenTech, Inc. Founded (2010)
  11. Pellionisz on the Board of Advisers in the USA and India (2011)
  12. ENCODE – final admission (2012)
  13. Recursive Genome Function is Clogged by Fractal Defects in Hilbert-Curve (2012)
  14. Geometric Unification of Neuroscience and Genomics (2012)
  15. US Patent Office issues FractoGene 8,280,641 to Pellionisz (2012)

file:///C|/Documents_and_Settings/Andras/Desktop/The_FractoGene_Decade_cover_page.htm  2012.12.16. 12:36:55

When the human genome was first sequenced in June 2000, there were two pretty big surprises.

The first was that humans have only about 30,000-40,000 identifiable genes,

  • not the 100,000 or more many researchers were expecting.

The lower –and more humbling — number

  • means humans have just one-third
  • more genes than a common species of worm.

The second stunner was how much human genetic material — more than 90 percent —

  • is made up of what scientists were calling “junk DNA.”

The term was coined to describe similar but

  • not completely identical repetitive sequences of amino acids
    (the same substances that make genes),
  • which appeared to have no function or purpose.

The main theory at the time was that these apparently

  • non-working sections of DNA were
  • just evolutionary leftovers, much like our earlobes.

If biophysicist Andras Pellionisz is correct, genetic science

  • may be on the verge of yielding its third — and
  • by far biggest — surprise.

With a doctorate in physics, Pellionisz is the holder of Ph.D.’s

  • in computer sciences and experimental biology from the
    prestigious Budapest Technical University and
    the Hungarian National Academy of Sciences.

A biophysicist by training, the 59-year-old is a former research

  1. associate professor of physiology and biophysics at New York University,
  2. author of numerous papers in respected scientific journals and textbooks,
  3. a past winner of the prestigious Humboldt Prize for scientific research,
  4. a former consultant to NASA and
  5. holder of a patent on the world’s first artificial cerebellum,
    a technology that has already been integrated into research
    on advanced avionics systems.

Because of his background, the Hungarian-born brain researcher might

  • also become one of the first people to successfully launch a new company
  • by using the Internet to gather momentum for a novel scientific idea.

The genes we know about today, Pellionisz says, can be thought of as something

  • similar to machines that make bricks (proteins, in the case of genes), with certain
  • junk-DNA sections providing a blueprint for the
  • different ways those proteins are assembled.

The notion that at least certain parts of junk DNA might have a purpose for example,

  • many researchers now refer to
  • with a far less derogatory term: introns.

Insert picture

3-d-genome-map

3-d-genome-map

In a provisional patent application filed July 31, Pellionisz claims to have

  • unlocked a key to the hidden role junk DNA plays in growth — and in life itself.

His patent application covers all attempts to

  • count,
  • measure and
  • compare

the fractal properties of introns

  • for diagnostic and therapeutic purposes.

IIIB. The Hidden Fractal Language of Intron DNA

To fully understand Pellionisz’ idea,

  • one must first know what a fractal is.

Fractals are a way that nature organizes matter.

Fractal patterns can be found

  • in anything that has a nonsmooth surface (unlike a billiard ball),
  1. such as coastal seashores,
  2. the branches of a tree or
  3. the contours of a neuron (a nerve cell in the brain).

Some, but not all, fractals are self-similar and

  • stop repeating their patterns at some stage

the branches of a tree, for example,

  • can get only so small.

Because they are geometric, meaning they have a shape,

  • fractals can be described in mathematical terms.

It’s similar to the way a circle can be described

  • by using a number to represent its radius
    (the distance from its center to its outer edge).

When that number is known, it’s possible to draw the circle it represents

  • without ever having seen it before.

Although the math is much more complicated,

  • the same is true of fractals.

If one has the formula for a given fractal,

  • it’s possible to use that formula to construct, or reconstruct,
  • an image of whatever structure it represents,
  • no matter how complicated.

The mysteriously repetitive but not identical strands of genetic material

  • are in reality building instructions organized in
  • a special type of pattern known as a fractal.

It’s this pattern of fractal instructions, he says, that tells genes what they

  • must do in order to form living tissue,
  • everything from the wings of a fly to the entire body of a full-grown human.

In a move sure to alienate some scientists,

  • Pellionisz has chosen the unorthodox route of
  • making his initial disclosures online on his own Web site.

He picked that strategy, he says, because

  1. it is the fastest way he can document his claims
  2. and find scientific collaborators and investors.

Most mainstream scientists usually blanch at such approaches,

  • preferring more traditionally credible methods, such as
  • publishing articles in peer-reviewed journals.

Basically, Pellionisz’ idea is that

  • a fractal set of building instructions in the DNA
  • plays a similar role in organizing life itself.

Decode the way that language works, he says, and

  • in theory it could be reverse engineered.

Just as knowing the radius of a circle lets one create that circle,

  • the more complicated fractal-based formula
  • would allow us to understand how nature creates a heart or
  • simpler structures, such as disease-fighting antibodies.

At a minimum, we’d get a far better understanding of

  • how nature gets that job done.

The complicated quality of the idea is helping encourage

  • new collaborations across the boundaries that sometimes
  • separate the increasingly intertwined disciplines of
  • biology, mathematics and computer sciences.

Hal Plotkin, Special to SF Gate. Thursday, November 21, 2002.

http://www.junkdna.com/plotkin.htm

(1 of 10)2012.12.13. 12:11:58/ Hal Plotkin, Special to SF Gate.
Thursday, November 21, 2002

insert pictures

Hilbert3d

Hilbert3d

Hilbert512

Hilbert512

Fractal Defects in the genome, repeat structural variants withtheir largest example of Copy Number Variants

Fractal Defects in the genome, repeat structural variants with their largest example of Copy Number Variants

Golden_ratio  Fractal chaos Holographic neural network

Golden_ratio Fractal chaos Holographic neural network

IIIC. multifractal analysis

The human genome: a multifractal analysis.
Moreno PA, Vélez PE, Martínez E, et al. BMC Genomics 2011, 12:506.

http://www.biomedcentral.com/1471-2164/12/506

Background: Several studies have shown that genomes

  • can be studied via a multifractal formalism.

Recently, we used a multifractal approach to study the

  • genetic information content of the Caenorhabditis elegans genome.

Here we investigate the possibility that the human genome shows a

  • similar behavior to that observed in the nematode.

Results: We report here multifractality in the human genome sequence.

This behavior correlates strongly on the presence of

  1. Alu elements and to a lesser extent on
  2. CpG islands and (G+C) content.

In contrast, no or low relationship was found for

  • LINE, MIR, MER, LTRs elements and DNA regions
  • poor in genetic information.

Gene function, cluster of orthologous genes, metabolic pathways, and exons

  1. tended to increase their frequencies with ranges of multifractality
  2. and large gene families were located in genomic regions with varied multifractality.

Additionally, a multifractal map and classification for human chromosomes are proposed.

Conclusions: we propose a descriptive non-linear model

for the structure of the human genome,

This model reveals a multifractal regionalization where

many regions coexist that are far from equilibrium and

this non-linear organization has significant molecular and medical genetic implications

  • for understanding the role of Alu elements in genome stability
  • and structure of the human genome.

Given the role of Alu sequences in

  1. adaptation and
  2. human genetic diversity,
  3. genetic diseases,
  4. gene regulation,
  5. phylogenetic analyses,

these quantifications are especially useful.

MiIP:The Monomer Identification and Isolation Program

Bun C, Ziccardi W, Doering J and Putonti C.
Evolutionary Bioinformatics 2012:8 293-300.
http://dx.doi.org:/10.4137/EBO.S9248

Repetitive elements within genomic DNA are

  • both functionally and evolutionarilly informative.

Discovering these sequences ab initio

  • is computationally challenging,
  • compounded by the fact that sequence identity
  • between repetitive elements can vary significantly.

Here we present a new application,

  • the Monomer Identification and Isolation Program (MiIP),
  • which provides functionality to both
  1. search for a particular repeat
  2. as well as discover repetitive elements within a larger genomic sequence.

To compare MiIP’s performance with other repeat detection tools,

  • analysis was conducted for synthetic sequences as well as
  • several a21-II clones and HC21 BAC sequences.

The primary benefit of MiIP is the fact that

  1. it is a single tool capable of searching for both known monomeric sequences
  2. as well as discovering the occurrence of repeats ab initio,
  3. per the user’s required sensitivity of the search

Triplex DNA A. A third strand for DNA

The DNA double helix can under certain conditions

  • accommodate a third strand in its major groove.

Researchers in the UK have now presented a complete set of

  • four variant nucleotides that makes it possible to use this phenomenon
  • in gene regulation and mutagenesis.

Natural DNA only forms a triplex

  • if the targeted strand is rich in purines – guanine (G) and adenine (A) –
  • which in addition to the bonds of the Watson-Crick base pairing
  • can form two further hydrogen bonds, and the ‘third strand’ oligonucleotide
  • has the matching sequence of pyrimidines – cytosine (C) and thymine (T).

Any Cs or Ts in the target strand of the duplex will only bind very weakly,

  • as they contribute just one hydrogen bond.

Moreover, the recognition of G requires

  • the C in the probe strand to be protonated,
  • so triplex formation will only work at low pH.

To overcome all these problems, the groups of Tom Brown and Keith Fox
at the University of Southampton

  • have developed modified building blocks, and have now
  • completed a set of four new nucleotides, each of which will bind to one
  • DNA nucleotide from the major groove of the double helix.1

They tested the binding of a 19-mer of these designer nucleotides

  • to a double helix target sequence in comparison with the corresponding
  • triplex-forming oligonucleotide made from natural DNA bases.

Using fluorescence-monitored thermal melting and DNase I footprinting,

  • the researchers showed that their construct
  • forms stable triplex even at neutral pH.

Tests with mutated versions of the target sequence showed that

  1. three of the novel nucleotides are highly selective for their target base pair,
  2. while the ‘S’ nucleotide, designed to bind to T, also tolerates C.

In principle, triplex formation has already been demonstrated as

  • a way of inducing mutations in cell cultures and animal experiments.2

Michael Gross

References

1 DA Rusling et al, Nucleic Acids Res. 2005, 33, 3025

http://NucleicAcidsRes.com/Rusling_DA

2 KM Vasquez et al, Science 2000, 290, 530

http://Science.org/Vazquez_KM

B. Triplex DNA Structures.

Triplex DNA Structures. Frank-Kamenetskii, Mirkin SM. Annual Rev Biochem 1995; 64:69-95./ www.annualreviews.org/aronline

Since the pioneering work of Felsenfeld, Davies, & Rich (1),

  • double-stranded polynucleotides containing purines in one strand
  • and pydmidines in the other strand
    [such as poly(A)/poly(U), poly(dA)/poly(dT), or poly(dAG)/poly(dCT)]
  • have been known to be able to undergo a
  • stoichiometric transition forming a triple-stranded structure containing
  • one polypurine and two polypyrimidine strands.

Early on, it was assumed that the third strand was located in the major groove

  • and associated with the duplex via non-Watson-Crick interactions
  • now known as Hoogsteen pairing.

Insert pictures

triplex DNA

triplex DNA

Triple helices consisting of one pyrimidine and

  • two purine strands were also proposed.

However, notwithstanding the fact that single-base triads

  1. in tRNAs tructures were well-documented,
  2. triple-helical DNA escaped wide attention before the mid-1980s.

The considerable modern interest in DNA triplexes arose

  • due to two partially independent developments.

First, homopurine-homopyrimidine stretches in supercoiled plasmids

  • were found to adopt an unusual DNA structure, called H-DNA which
  • includes a triplex as the major structural element.

Secondly, several groups demonstrated that homopyrimidine and

  • some purine-rich oligonucleotides
  • can form stable and sequence-specific complexes
  • with corresponding homopurine-homopyrimidine sites on duplex DNA.

These complexes were shown to be triplex structures rather than D-loops,

  • where the oligonucleotide invades the double helix
  • and displaces one strand.

A characteristic feature of all these triplexes is that the two chemically

  • homologous strands (both pyrimidine or both purine) are antiparallel.

These findings led explosive growth in triplex studies. One can easily imagine

  • numerous “geometrical” ways to form a triplex, and
  • those that have been studied experimentally.

The canonical intermolecular triplex consists of either

  1. three independent oligonucleotide chains or of
  2. a long DNA duplex carrying homopurine-homopyrimidine insert
  • and the corresponding oligonucleotide.

Triplex formation strongly depends on the oligonucleotide(s) concentration.

A single DNA chain may also fold into a triplex connected by two loops.

To comply with the sequence and polarity requirements for triplex formation,

  • such a DNA strand must have a peculiar sequence:

It contains a mirror repeat
(homopyrimidine for YR*Y triplexes and homopurine for YR*R triplexes)

  • flanked by a sequence complementary to
  • one half of this repeat.

Such DNA sequences fold into

  • triplex configuration much more readily than do
  • the corresponding intermolecular triplexes, because
  • all triplex forming segments are brought together within the same molecule.

Insert pictures

It has become clear recently, however, that

  • both sequence requirements and chain polarity rules for triplex formation
  • can be met by DNA target sequences
  • built of clusters of purines and pyrimidines.

The third strand consists of adjacent homopurine and homopyrimidine blocks

  • forming Hoogsteen hydrogen bonds with purines
  • on alternate strands of the target duplex, andthis strand switch
  • preserves the proper chain polarity.

These structures, called alternate-strand triplexes,

  • have been experimentally observed as both intra- and intermolecular triplexes.

These results increase the number of

  • potential targets for triplex formation in natural DNAs
  • somewhat by adding sequences composed of purine and pyrimidine clusters,
  • although arbitrary sequences are still not targetable
  • because strand switching is energetically unfavorable.

References:

Lyamichev VI, Mirkin SM, Frank-Kamenetskii MD.

J. Biomol. Stract. Dyn. 1986; 3:667-69.

http://JbiomolStractDyn.com/Lyamichev_VI/

Mirkin SM, Lyamichev VI, Drushlyak KN, Dobrynin VN0 Filippov SA, Frank-Kamenetskii MD.

Nature 1987; 330:495-97.

http://Nature.com/

Demidov V, Frank-Kamenetskii MD, Egholm M, Buchardt O, Nielsen PE.

Nucleic Acids Res. 1993; 21:2103-7.

http://NucleicAcidsResearch.com/

Mirkin SMo Frank-Kamenetskii MD.

Anna. Rev. Biophys. Biomol. Struct. 1994; 23:541-76.

http://AnnRevBiophysBiomolecStructure.com/

Hoogsteen K.

Acta Crystallogr. 1963; 16:907-16

http://ActaCrystallogr.com/

Malkov VA, Voloshin ON, Veselkov AG, Rostapshov VM, Jansen I, et al.

Nucleic Acids Res. 1993; 21:105-11.

http://NucleicAidsResearch.com/

Malkov VA, Voloshin ON, Soyfer VN, Frank-Kamenetskii MD.

Nucleic Acids Res. 1993; 21:585-91

Chemy DY, Belotserkovskii BP,Frank-Kamenetskii MD,
Egholm M, Buchardt O, et al.

Proc. Natl. Acad. Sci. USA 1993; 90:1667-70

http://PNAS.org/

C.Triplex forming oligonucleotides

Triplex forming oligonucleotides: sequence-specific tools for genetic targeting.

Knauert MP, Glazer PM. Human Molec Genetics 2001; 10(20):2243-2251. http://HumanMolecGenetics.com/Triplex_forming_oligonucleotides:
sequence-specific_tools_for _genetic_targeting.

Triplex forming oligonucleotides (TFOs) bind in the major groove of duplex DNA

  • with a high specificity and affinity.

Because of these characteristics,

  • TFOs have been proposed as homing devices
  • for genetic manipulation in vivo.

These investigators review work demonstrating the ability of TFOs and

  • related molecules to alter gene expression and
  • mediate gene modification in mammalian cells.

TFOs can mediate targeted gene knock out in mice,

  • providing a foundation for potential application
  • of these molecules in human gene therapy.

D. Novagon DNA

John Allen Berger, founder of Novagon DNA and

  • The Triplex Genetic Code Over the past 12+ years,

Novagon DNA has amassed a vast array of empirical findings

  • which challenge the “validity” of the “central dogma theory”,
  • especially the current five nucleotide Watson-Crick DNA and
  • RNA genetic codes. DNA = A1T1G1C1, RNA =A2U1G2C2.

We propose that our new Novagon DNA 6 nucleotide Triplex Genetic Code

  • has more validity than the existing 5 nucleotide (A1T1U1G1C1)
  • Watson-Crick genetic codes.

Our goal is to conduct a “world class” validation study

  • to replicate and extend our findings.

Methods for Examining Genomic and Proteomic Interactions

A. An Integrated Statistical Approach to Compare
Transcriptomics Data Across Experiments:

A Case Study on the Identification of Candidate Target Genes
of the Transcription Factor PPARα

Ullah MO, Müller M and Hooiveld GJEJ.

Bioinformatics and Biology Insights 2012:6 145–154.

binding-of-a-ppar-ligand-to-the-ppar-ligand-binding-domain

binding-of-a-ppar-ligand-to-the-ppar-ligand-binding-domain

http://bionformaticsandBiologyInsights.com/An_Integrated_Statistical_Approach_to_Compare_ transcriptomic_Data_Across_Experiments-A-Case_Study_on_the_Identification_ of_Candidate_Target_Genes_of_the Transcription_Factor_PPARα/

Corresponding author email: guido.hooiveld@wur.nl

An effective strategy to elucidate the signal transduction cascades

  • activated by a transcription factor is to compare the transcriptional profiles
  • of wild type and transcription factor knockout models.

Many statistical tests have been proposed for analyzing gene expression data,

  • but most tests are based on pair-wise comparisons.

Since the analysis of micro-arrays involves the testing of

  • multiple hypotheses within one study, it is generally accepted that one should
  • control for false positives by the false discovery rate (FDR).

However, it has been reported that

  • this may be an inappropriate metric for
  • comparing data across different experiments.

Here we propose an approach that addresses the above mentioned problem

  • by the simultaneous testing and integration of the three hypotheses (contrasts)
  • using the cell means ANOVA model.

These three contrasts test for the effect of a treatment in

  • wild type,
  • gene knockout, and
  • globally over all experimental groups.

We illustrate our approach on microarray experiments that focused

  • on the identification of candidate target genes and biological processes
  • governed by the fatty acid sensing transcription factor PPARα in liver.

Compared to the often applied FDR based across experiment comparison,

  • our approach identified a conservative
  • but less noisy set of candidate genes
  • with same sensitivity and specificity.

However, our method had the advantage of properly adjusting for

  • multiple testing while integrating data from two experiments,
  • and was driven by biological inference.

We present a simple, yet efficient strategy to compare

  • differential expression of genes across experiments
  • while controlling for multiple hypothesis testing.

B. Managing biological complexity across orthologs with a visual knowledge-base
of documented biomolecular interactions Vincent VanBuren & Hailin Chen
Scientific Reports 2, Article number: 1011
http://dx.doi.org:/10.1038/srep01011
Received 02 October 2012 Accepted 04 December 2012

The complexity of biomolecular interactions and influences

  • is a major obstacle to their comprehension and elucidation.

Visualizing knowledge of biomolecular interactions

  • increases comprehension and
  • facilitates the development of new hypotheses.

The rapidly changing landscape of high-content experimental results

  • also presents a challenge for the maintenance of comprehensive knowledgebases.

Distributing the responsibility for maintenance of a knowledgebase

  • to a community of subject matter experts is an effective strategy
  • for large, complex and rapidly changing knowledgebases.

Cognoscente serves these needs by building visualizations for queries

  • of biomolecular interactions on demand,
  • by managing the complexity of those visualizations, and by
  • crowdsourcing to promote the incorporation of current knowledge
  • from the literature.

Imputing functional associations between

  • biomolecules and imputing directionality of regulation for those predictions
  • each require a corpus of existing knowledge as a framework to build upon.

Comprehension of the complexity of this corpus of knowledge

  • will be facilitated by effective visualizations of
  • the corresponding biomolecular interaction networks.

Cognoscente (http://vanburenlab.medicine.tamhsc.edu/cognoscente.html)

  1. was designed and implemented to serve these roles as a knowledgebase
  2. and as an effective visualization tool for systems biology research and education.

Cognoscente currently contains over 413,000 documented interactions,

  • with coverage across multiple species.

Perl, HTML, GraphViz1, and a MySQL database were used in the development of Cognoscente.

Cognoscente was motivated by the need to update the knowledgebase

  • of biomolecular interactions at the user level, and
  • flexibly visualize multi-molecule query results for
  • heterogeneous interaction types across different orthologs.

Satisfying these needs provides a strong foundation for

  • developing new hypotheses about regulatory and metabolic pathway topologies.

Several existing tools provide functions that are similar to Cognoscente, so we selected several popular alternatives to assess how their feature sets compare with Cognoscente ( Table 1 ). All databases assessed had easily traceable documentation for each interaction, and included protein-protein interactions in the database.

Most databases, with the exception of BIND, provide an open-access database that can be downloaded as a whole.

Most databases, with the exceptions of EcoCyc and HPRD, provide

  • support for multiple organisms.

Most databases support web services for

  • interacting with the database contents programmatically,
  • whereas this is a planned feature for Cognoscente.

INT, STRING, IntAct, EcoCyc, DIP and Cognoscente provide built-in

  • visualizations of query results, which we consider
  • among the most important features for facilitating comprehension of query results.

BIND supports visualizations via Cytoscape.

Cognoscente is among a few other tools that support

  • multiple organisms in the same query,
  • protein->DNA interactions, and
  • multi-molecule queries.

Cognoscente has planned support for

  • small molecule interactants (i.e. pharmacological agents).

MINT, STRING, and IntAct provide a prediction (i.e. score)

  • of functional associations, whereas
  • Cognoscente does not currently support this.

Cognoscente provides support for multiple edge encodings

  • to visualize different types of interactions in the same display,
  • a crowdsourcing web portal that allows users to submit
  • interactions that are then automatically incorporated in the knowledgebase,
  • and displays orthologs as compound nodes
  • to provide clues about potential orthologous interactions.

The main strengths of Cognoscente are that it provides a combined feature set that is superior to any existing database, it provides a unique visualization feature for orthologous molecules, and relatively unique support for multiple edge encodings, crowdsourcing, and connectivity parameterization. The current weaknesses of Cognoscente relative to these other tools are that it does not fully support web service interactions with the database, it does not fully support small molecule interactants, and it does not score interactions to predict functional associations. Web services and support for small molecule interactants are currently under development.

Related references from Leaders in Pharmaceutical Intelligence:

Big Data in Genomic Medicine larryhbern
http://pharmaceuticalintelligence.com/2012/12/17/big-data-in-genomic-medicine/

BRCA1 a tumour suppressor in breast and ovarian cancer – functions in
transcription, ubiquitination and DNA repair
S Saha
http://pharmaceuticalintelligence.com/2012/12/04/brca1-a-tumour-suppressor-in-breast-and-ovarian-cancer-functions-in-transcription-ubiquitination-and-dna-repair/

Computational Genomics Center: New Unification of Computational Technologies at Stanford
A Lev-Ari    http://pharmaceuticalintelligence.com/2012/12/03/computational-genomics-center-new-unification-of-computational-technologies-at-stanford/

Personalized medicine gearing up to tackle cancer
ritu saxena
http://pharmaceuticalintelligence.com/2013/01/07/personalized-medicine-gearing-up-to-tackle-cancer/

Differentiation Therapy – Epigenetics Tackles Solid Tumors

SJ Williams   http://pharmaceuticalintelligence.com/2013/01/03/differentiation-therapy-epigenetics-tackles-solid-tumors/

Mechanism involved in Breast Cancer Cell Growth: Function in Early Detection & Treatment
A Lev-Ari
http://pharmaceuticalintelligence.com/2013/01/17/mechanism-involved-in-breast-cancer-cell-growth-function-in-early-detection-treatment/

The Molecular pathology of Breast Cancer Progression
tilde barliya
http://pharmaceuticalintelligence.com/2013/01/10/the-molecular-pathology-of-breast-cancer-progression/

Gastric Cancer: Whole-genome reconstruction and mutational signatures
A Lev-Ari
http://pharmaceuticalintelligence.com/2012/12/24/gastric-cancer-whole-genome-reconstruction-and-mutational-signatures-2/

Paradigm Shift in Human Genomics – Predictive Biomarkers and Personalized Medicine – Part 1 (pharmaceuticalintelligence.com)
A Lev-Ari
http://pharmaceuticalintelligence.com/2013/01/13/paradigm-shift-in-human-genomics-predictive-biomarkers-and-personalized-medicine-part-1/

LEADERS in Genome Sequencing of Genetic Mutations for Therapeutic Drug Selection in Cancer Personalized Treatment: Part 2
A Lev-Ari
http://pharmaceuticalintelligence.com/2013/01/13/leaders-in-genome-sequencing-of-genetic-mutations-for-therapeutic-drug-selection-in-cancer-personalized-treatment-part-2/

Personalized Medicine: An Institute Profile – Coriell Institute for Medical Research: Part 3
A Lev-Ari
http://pharmaceuticalintelligence.com/2013/01/13/personalized-medicine-an-institute-profile-coriell-institute-for-medical-research-part-3/

Harnessing Personalized Medicine for Cancer Management, Prospects of Prevention and Cure: Opinions of Cancer Scientific Leaders @ http://pharmaceuticalintelligence.com
A Lev-Ari
http://pharmaceuticalintelligence.com/2013/01/13/7000/Harnessing_Personalized_Medicine_for_ Cancer_Management-Prospects_of_Prevention_and_Cure/

GSK for Personalized Medicine using Cancer Drugs needs Alacris systems biology model to determine the in silico effect of the inhibitor in its “virtual clinical trial”
A Lev-Ari
http://pharmaceuticalintelligence.com/2012/11/14/gsk-for-personalized-medicine-using-cancer-drugs-needs-alacris-systems-biology-model-to-determine-the-in-silico-effect-of-the-inhibitor-in-its-virtual-clinical-trial/

Recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes in serous endometrial tumors
S Saha
http://pharmaceuticalintelligence.com/2012/11/19/recurrent-somatic-mutations-in-chromatin-remodeling-and-ubiquitin-ligase-complex-genes-in-serous-endometrial-tumors/

Personalized medicine-based cure for cancer might not be far away ritu saxena    http://pharmaceuticalintelligence.com/2012/11/20/personalized-medicine-based-cure-for-cancer-might-not-be-far-away/

Human Variome Project: encyclopedic catalog of sequence variants indexed to the human genome sequence A Lev-Ari http://pharmaceuticalintelligence.com/2012/11/24/human-variome-project-encyclopedic-catalog-of-sequence-variants-indexed-to-the-human-genome-sequence/

Prostate Cancer Cells: Histone Deacetylase Inhibitors Induce Epithelial-to-Mesenchymal Transition
SJ Williams
http://pharmaceuticalintelligence.com/2012/11/30/histone-deacetylase-inhibitors-induce-epithelial-to-mesenchymal-transition-in-prostate-cancer-cells/

Inspiration From Dr. Maureen Cronin’s Achievements in Applying Genomic Sequencing to Cancer Diagnostics A Lev-Ari http://pharmaceuticalintelligence.com/2013/01/10/inspiration-from-dr-maureen-cronins-achievements-in-applying-genomic-sequencing-to-cancer-diagnostics/

The “Cancer establishments” examined by James Watson, co-discoverer of DNA w/Crick, 4/1953
A Lev-Ari  http://pharmaceuticalintelligence.com/2013/01/09/the-cancer-establishments-examined-by-james-watson-co-discover-of-dna-wcrick-41953/

Directions for genomics in personalized medicine larryhbern
http://pharmaceuticalintelligence.com/2013/01/27/directions-for-genomics-in-personalized-medicine/

How mobile elements in “Junk” DNA promote cancer. Part 1: Transposon-mediated tumorigenesis. SJ Williams
http://pharmaceuticalintelligence.com/2012/10/31/how-mobile-elements-in-junk-dna-prote-cancer-part1-transposon-mediated-tumorigenesis/ Mitochondria: More than just the “powerhouse of the cell” ritu saxena  http://pharmaceuticalintelligence.com/2012/07/09/mitochondria-more-than-just-the-powerhouse-of-the-cell/

Mitochondrial fission and fusion: potential therapeutic targets? Ritu saxena    http://pharmaceuticalintelligence.com/2012/10/31/mitochondrial-fission-and-fusion-potential-therapeutic-target/

Mitochondrial mutation analysis might be “1-step” away ritu saxena  http://pharmaceuticalintelligence.com/2012/08/14/mitochondrial-mutation-analysis-might-be-1-step-away/

mRNA interference with cancer expression larryhbern
http://pharmaceuticalintelligence.com/2012/10/26/mrna-interference-with-cancer-expression/

Expanding the Genetic Alphabet and linking the genome to the metabolome http://pharmaceuticalintelligence.com/2012/09/24/expanding-the-genetic-alphabet-and-linking-the-genome-to-the-metabolome/

Breast Cancer, drug resistance, and biopharmaceutical targets larryhbern
http://pharmaceuticalintelligence.com/2012/09/18/breast-cancer-drug-resistance-and-biopharmaceutical-targets/

Breast Cancer: Genomic profiling to predict Survival: Combination of Histopathology and Gene Expression Analysis
A Lev-Ari
http://pharmaceuticalintelligence.com/2012/12/24/breast-cancer-genomic-profiling-to-predict-survival-combination-of-histopathology-and-gene-expression-analysis/

Gastric Cancer: Whole-genome reconstruction and mutational signatures A Lev-Ari http://pharmaceuticalintelligence.com/2012/12/24/gastric-cancer-whole-genome-reconstruction-and-mutational-signatures-2/

Ubiquinin-Proteosome pathway, autophagy, the mitochondrion, proteolysis and cell apoptosis larryhbern  http://pharmaceuticalintelligence.com/2012/10/30/ubiquinin-proteosome-pathway-autophagy-the-mitochondrion-proteolysis-and-cell-apoptosis/

Genomic Analysis: FLUIDIGM Technology in the Life Science and Agricultural Biotechnology A Lev-Ari http://pharmaceuticalintelligence.com/2012/08/22/genomic-analysis-fluidigm-technology-in-the-life-science-and-agricultural-biotechnology/

2013 Genomics: The Era Beyond the Sequencing Human Genome: Francis Collins, Craig Venter, Eric Lander, et al.  http://pharmaceuticalintelligence.com/2013_Genomics

Paradigm Shift in Human Genomics – Predictive Biomarkers and Personalized Medicine – Part 1 http://pharmaceuticalintelligence.com/Paradigm Shift in Human Genomics_/

Related Articles Life Stands on the shoulders of Giants (Viruses) (bytesizebio.net)

New insights into the human genome by ENCODE project (slideshare.net)

Unraveling the Human Genome: 6 Molecular Milestones (livescience.com)

Melanoma Genes Found In “Junk” DNA (medicalnewstoday.com)

Learning the alphabet of gene control (esciencenews.com)

BEST OF THE WEB: On viral ‘junk’ DNA, a DNA-enhancing Ketogenic diet, and cometary kicks (sott.net)

Sohan Modak

Sohan

Sohan Modak

Owner, Open vision Inc.

Top Contributor

Larry, in a series of papers, Fertil, Deschavannes and colleagues have done beautiful analyses of fractal diagrams of Genome sequences in a series of papers.[Deschavanne PJ, Giron A, Vilain J, Fagot G, Fertil B (1999) Mol Biol Evol 16: 1391-1399; Fertil B, Massin M, Lespinats S, Devic C, Dumee P, Giron A (2005) GENSTYLE: exploration and analysis of DNA sequences with genomic signature. Nucleic Acids Res 33(Web Server issue):W512-5]. Clearly this gives an extraordinary insight in the specificity of positional sequence clusters. While fractals work well with octanucleotide clusters, longer the oligonucleotide tracks, higher the resolution. I feel that high resolution fractal maps of fentanucleotide sequences will provide something truely different and may be used as a tool to compare normal cellular DNA sequences to those from cancer cell lines and provide an operational window for manipulations.

Read Full Post »

This is another notable post on the improvement in absorption and bioactivty of a drug, with toxicity known.

Read Full Post »

Another informative post of what goes into pharmaceutical analysis.

Larry H Bernstein, MD, FCAP

Read Full Post »

Interesting explanation of improvement in an existing drug.

Larry H Bernstein, MD, FCAP

Read Full Post »

The Human Proteome Map Completed

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

 

Researchers Produce First Map of Human Proteome, and Reveal New
Significance in The Human Proteome

HAHNE, TECHNISCHE UNIVERSITÄT MÜNCHENTwo international teams have
independently produced the first drafts of the human proteome. These curated
catalogs of the proteins expressed in most non-diseased human tissues and
organs can be used as a baseline to better understand changes that occur in
disease states. Their findings were published today (May 29) in Nature.

Both teams uncovered new complexities of the human genome, identifying novel
proteins from regions of the genome previously thought to be non-coding.

“the real breakthrough with these two projects is the comprehensive coverage of
more than 80 percent of the expected human proteome” said Hanno Steen, director
of proteomics at Boston Children’s Hospital, who was not involved in the work.

The human proteome map provides a catalog of proteins expressed in nondiseased tissues and organs to use as baseline in understanding changes that occur in disease

Given the growing importance of proteins in medical laboratory testing,

Experts are comparing this to the first complete map of the human genome

  • and this information provides for rapid advances
  • in understanding transcriptomics and metabolomics

Map of Human Proteome Expected to Advance Medical Science

“Housekeeping genes” that are expressed in all tissues and cell types

  • have been thought to be involved in basic cellular functions.

Two teams developing a Human Proteome Map

  • detected proteins encoded by 2,350 genes
  • across all human cells and tissues.

The corresponding housekeeping proteins comprised
about 75% of total protein mass.

  •  histones,
  • ribosomal proteins,
  • metabolic enzymes, and
  • cytoskeletal proteins

The two international teams produced

  • the first drafts of the human protoeome,
  • a catalog of proteins expressed in most
  • nondiseased human issues and organs.

The evidence suggests there is translation from DNA regions

  • that were not thought to be translated—including
  • more than 400 translated long, intergenic non-coding RNAs (lincRNAs)—
    found by the Küster team—and
  • 193 new proteins—uncovered by the Pandey team.

This proteome map can be used as a baseline to understand

  • changes that occur in the disease state

These studies are part of the Human Proteome Project,

  1. an international effort by the Human Proteome Organization
  2. to revolutionize our understanding of the human proteome
  3. by coordinating research at laboratories around the world directed
  4. at mapping the entire human proteome.

This new information about the human proteome

  • is expected to trigger rapid advances in medical science
  • and a better understanding of the underlying causes of human diseases.

One Study Team Was at Johns Hopkins University

  • In one study, which was headed by Ahilesh Pandey, M.D.,
    at Johns Hopkins University in Baltimore,
  • and colleague Harsha Gowda, Ph.D.,
    of the Institute of Bioinformatics in Bangalore, India,
  • the research team used an advanced form of mass spectrometry to analyze proteins
  • to create the human proteome map,

according to a report published in NIH Research Matters.

The research team examined

  1. 30 normal human tissue and cell types:
  2. 17 adult tissues,
  3. 7 fetal tissue and
  4. 6 blood cell types.

Samples from three people per tissue type

  • were processed through several steps.

The protein fragments, or peptides, were analyzed on

The amino acid sequences were

  • then compared to known sequences.

Their results were published in the May 28, 2014, issue of Nature.

The resulting draft map of the human proteome map includes

  • proteins encoded by more than 17,000 genes,
  • noted the Research Matters article.

Among these are hundreds of proteins from regions

  • previously thought to be non-coding.

This study also provided a new understanding of

  • how genes are expressed.

For example, almost 200 genes begin in locations

  • other than those predicted based on genetic sequence.

“The fact that 193 of the proteins came from DNA sequences

  • predicted to be non-coding means that
  • we don’t fully understand how cells read DNA,
  • since the sequences code for proteins

This study also produced the Human Proteome Map,

  • an interactive online portal.

This can be accessed at this link.

The study data will soon be accessible through

German’s ProteomicsDB Analyzed a Mix of Available and New Tissue Data

The other study was conducted by a team lead by  Bernhard Küster
of the Technische Universität München in Germany.

Küster and his colleagues created a

This database contains 92% of the

  • estimated 19,629 human proteins,

noted The Scientist article.

Küster’s team also used mass spectrometry

  • to analyze human tissue samples.

This team’s approach differed from Johns Hopkins’ in that

  • it compiled about 60% of the information
  • in the ProteomicsDB database
  1. by using existing raw mass spec (MS) data
  2. from databases and colleagues’ contributions.

To fill data gaps, the Küster lab generated its own
MS data after analyzing

  1. 60 human tissues,
  2. 13 body fluids, and
  3. 147 cancer cell lines.

High-resolution public data

  • was selected and computationally processed
  • for strict quality

The database for ProteomicsDB is

  • public and searchable.

It can be accessed at this link.

German Study Added New Insights to Transcription Process

Comparing the ratio of protein to mRNA levels for every protein globally,

  • the Küster lab found that the translation rate
  • is a constant feature of each mRNA transcript. 

The proteomics community has viewed

  • transcriptome and proteome data as two sides of a coin.

But this analysis shows that at least, at steady state,

  • once the ratio for an mRNA/protein pair has been calculated,
  1. protein levels can be determined
  2. just from specific mRNA levels.

Proteomics researchers in Toronto maintaining ionic balance and in Boston commented on the
importance of the findings, even a “new paradigm” because of

  • the fixed ratio of protein to mRNA

This is quite in keeping with what we have been learning

  • with respect to homeostasis.

In 2003, the Human Genome Project created a

  • draft map of the human genome—
  • all the genes in the human body.

Genomics has since driven many advances in medical science.

This was a progress from the classic discovery of Watson and Crick –

  • the classical dogma holds that
  • DNA makes RNA makes protein.
  • no constraints are place on this

But the cell is functioning in contact with other cells,

  • immersed in interstitial fluid
  • maintaining cationic and anionic balance
  • and mitochondrial energy balance and ubiquitin systems interact
  • and protein interacts with the chromatin and transcriptional RNA

So the restriction that has been discovered has credence,

  • the classical diagram has to be redrawn

Deeper Knowledge of Proteome to Improve Diagnostics and Therapeutics

In the two projects is:

  • the comprehensive coverage of more than 80% of
  • the expected human proteome,

These studies indicate that to get to

  • a deep level of proteome coverage,
  • many different tissue types must be probed.

the  studies are  complimentary.

  1. The Hopkins group provided a survey of human proteins from a single source, which allows for easy comparisons within their data.
  2. The ProteomeDB effort connected new information with existing data

A deeper knowledge of the human proteome could help

  • fill the gap between genomes and phenotypes.

As this occurs, it has the potential to transform

  • the way diagnostics and therapeutics are developed,
  •  enhancing overall biomedical research and healthcare,

it was noted in a report presented to scientific leaders at a NIH workshop

  • on advances in proteomics and its applications.

Having completed a draft map of the human proteome—
the set of all proteins in the human body

  • It opens another window to cell function.

It has been ASSUMED –

  • genes control the most basic functions of the cell,
  • including what proteins to make and when.
  • but we have assumed for too much in assigning
    full control to the genome

Researchers have identified more than 20,000 protein- coding genes.

However, scientific understanding of the proteome has

  • lagged behind that of the genome,
  • partly because of the proteome’s complexities.

The relationship between genes and proteins isn’t a simple matter of

  • one gene coding for one protein.

Stretches of DNA can be read and translated

  • into proteins in different ways.

Proteins are also more difficult to sequence than genes.

The importance of these latest studies to pathologists and Ph.D.s working

  • in molecular diagnostics laboratories is that
  • this information will expedite further research into the human proteome.

Such research is expected to lead to

  • novel methods of diagnosis and complex
  • “multi-analyte” clinical laboratory tests that
  • look for multiple proteins in a single assay.

“The prevalent view was that information transfer was from genome to transcriptome to proteome.
What these efforts show is that it’s a two-way road— proteomics can be used to annotate the genome.
The importance is that, using these datasets, we can improve the annotation of the genome and the
algorithms that predict transcription and translation,” said Steen. “The genomics field can now hugely
benefit from proteomics data.”

Wilhelm et al., “Mass-spectrometry- based draft of the human proteome,”
Nature,  http://dx.doi.doi:/10.1038/nature13319, 2014

M.S. Kim et al. “A draft map of the human proteome,”
Nature,  http://dx.doi.org:/10.1038/nature13302, 2014.

Tags

proteomicsnoncoding RNAhuman researchhuman proteome projecthuman genetics and genomics

http://www.the-scientist.com/?articles.view/articleNo/40083/title/Human-Proteome-Mapped/

 

__Patricia Kirk

__by Harrison Wein, Ph.D.

__by Anna Azvolinsky

Related Information:

Revealing The Human Proteome

Human Proteome Mapped

The human proteome – a scientific opportunity for transforming diagnostics, therapeutics, and healthcare

Reference: A draft map of the human proteome.
Kim MS, Pinto SM, Getnet D, Nirujogi RS, Manda SS, Donahue CA, Gowda H, Pandey A.
Nature. 2014 May 29;509(7502):575-81. http://dx.doi.org:/10.1038/nature13302. PMID: 24870542

Funding: NIH’s National Institute of General Medical Sciences (NIGMS), National Cancer Institute (NCI),
and National Heart, Lung, and Blood Institute (NHLBI); the Sol Goldman Pancreatic Cancer Research Center;
India’s Council of Scientific and Industrial Research; and Wellcome Trust/DBT India Alliance.

http://nihprod.cit.nih.gov/researchmatters/june2014/06092014proteome.htm

 

 

 

 

 

 

 

 

 

Read Full Post »

New Frontiers in Gene Editing — Cambridge Healthtech Institute’s Inaugural, February 19-20, 2015 | The Inter Continental San Francisco | San Francisco, CA

Reporter: Aviva Lev-Ari, PhD, RN

Cambridge Healthtech Institute’s Inaugural

New Frontiers in Gene Editing

Transitioning From the Lab to the Clinic

February 19-20, 2015 | The InterContinental San Francisco | San Francisco, CA
Part of the 22nd International Molecular Medicine Tri-Conference

 

Gene editing is rapidly progressing from being a research/screening tool to one that promises important applications downstream in drug development and cell therapy. Cambridge Healthtech Institute’s inaugural symposium on New Frontiers in Gene Editing will bring together experts from all aspects of basic science and clinical research to talk about how and where gene editing can be best applied. What are the different tools that can be used for gene editing, and what are their strengths and limitations? How does the CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas system, compare to Transcription Activator-like Effector Nucleases (TALENs), zinc finger nucleases (ZFNs) and other systems and where are they being used? Scientists and clinicians from pharma/biotech as well as from academic and government labs will share their experiences leveraging the utility of gene editing for functional screening, creating cell lines and knock-outs for disease modeling, and for cell therapy.

 

KEYNOTE PRESENTATIONS:

Precise Single-Base Genome Engineering for Human Diagnostics and Therapy

Bruce R. Conklin M.D., Investigator, Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes and Professor, Division of Genomic Medicine, University of California, San Francisco

Genome Edited Induced Pluripotent Stem Cells for Drug Screening

Joseph C. Wu, M.D., Ph.D., Director, Stanford Cardiovascular Institute and Professor, Department of Medicine/Cardiology & Radiology, Stanford University School of Medicine

 

USING GENE EDITING FOR FUNCTIONAL SCREENS

Exploration of Cellular Stress and Trafficking Pathways Using shRNA and CRISPR/Cas9-based Systems

Michael Bassik, Ph.D., Assistant Professor, Department of Genetics, Stanford University

Gene Editing in Patient-derived Stem Cells for In Vitro Modeling of Parkinson’s Disease

Birgitt Schuele M.D., Associate Professor and Director of Gene Discovery and Stem Cell Modeling, The Parkinson’s Institute

Massively Parallel Combinatorial Genetics to Overcome Drug Resistance in Bacterial Infections and Cancer

Timothy K. Lu, M.D., Ph.D., Associate Professor, Synthetic Biology Group, Department of Electrical Engineering and Computer Science and Department of Biological Engineering, Synthetic Biology Center, Massachusetts Institute of Technology

 

TRANSLATING GENE EDITING IN VIVO

CRISPR-Cas: Tools and Applications for Genome Editing

Fei Ann Ran, Ph.D., Post-doctoral Fellow, Laboratory of Dr. Feng Zhang, Broad Institute and Junior Fellow, Harvard Society of Fellows

Anti-HIV Therapies: Genome Engineering the Virus and the Host

Paula M. Cannon Ph.D., Associate Professor, Molecular Microbiology & Immunology, Biochemistry, and Pediatrics, Keck School of Medicine, University of Southern California

Preventing Transmission of Mitochondrial Diseases by Germline Heteroplasmic Shift Using TALENs

Juan Carlos Izpisua Belmonte, Ph.D., Professor, Gene Expression Laboratory, Salk Institute

Nuclease-Based Gene Correction for Treating Single Gene Disorders

Gang Bao, Ph.D., Professor, Robert A. Milton Chair in Biomedical Engineering, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University

 

EXPLORING GENE EDITING FOR THERAPEUTIC USES

Gene Editing on the Cusp of Exciting Opportunities for Human Therapeutics

Rodger Novak, M.D., CEO, CRISPR Therapeutics

Genome Editing for Genetic Diseases of the Blood

Matthew Porteus, M.D., Ph.D., Associate Professor, Pediatrics, Stanford University School of Medicine

Genome Engineering Tools for Gene Therapy and Regenerative Medicine

Charles A. Gersbach, Ph.D., Assistant Professor, Department of Biomedical Engineering, Center for Genomic and Computational Biology, Duke University

 

INTELLECTUAL PROPERTY LANDSCAPE: OPPORTUNITIES & CONCERNS

CRISPR/Cas-9: Navigating Intellectual Property (IP) Challenges in Gene Editing

Chelsea Loughran, Associate, Litigation Group, Wolf, Greenfield and Sacks, P.C.

Suggested Event Package:

February 15 Afternoon Short Course: Best Practices in Personalized and Translational Medicine
February 15 Dinner Short Course: Regulatory Compliance in Drug-Diagnostics Co-Development
February 16 Morning Short Course: Isolation and Characterization of Cancer Stem Cells
February 16-18 Conference Program: Genome and Transcriptome Analysis

 

 

For more details on the conference, please contact: 
Tanuja Koppal, Ph.D.,
Conference Director
Cambridge Healthtech Institute
E: tkoppal@healthtech.com

For partnering and sponsorship information, please contact: 
Jon Stroup (Companies A-K)
Manager, Business Development
Cambridge Healthtech Institute
T: (+1) 781-972-5483
E: jstroup@healthtech.com

Joseph Vacca (Companies L-Z)
Manager, Business Development
Cambridge Healthtech Institute
T: (+1) 781.972.5431
E: jvacca@healthtech.com

SOURCE

http://www.triconference.com/gene-editing

From: Gene Editing <davem@healthtech.com>
Date: Wed, 27 Aug 2014 12:58:56 -0400
To: <avivalev-ari@alum.berkeley.edu>
Subject: New Frontiers in Gene Editing [preliminary agenda just released]

Read Full Post »

Buffering of genetic modules involved in tricarboxylic acid cycle metabolism provides homeomeostatic regulation

Larry H. Bernstein, MD, FCAP, Reviewer and Curator

Leaders in Pharmaceutical Intelligence

This posting is the fifth in a series on metabolomics.  The first covered general principles.  Proteomics has not been covered and will be returned to.  But we have opened a door.  We have now looked at a comparison of two lymphocytic cell lines, and then the measurement of external effluxes to define internal metabolic conditions in yeast, with a view to delineating relationships between internal metabolic pathways and genetic variants under metabolic constraints.  These studies were confined to the experimental conditions, and could not measure metabolic fluxes, but I consider a study of the fluxes referred to in the comment by Dr. Jose Eduardo des Salles (JEDS) Roselino, who has brought up the concept, not yet specifically discussed – homeostasis.  It is part of a series of communications over several months.  

In the last article I might not have provided answers to some of the questions posed up front.  One of them refers to whether one finds a relationship to the Pasteur effect.  In both studies, leukemia cells and yeast, the cells are eukariotic, not prokaryotic, although the studies were preceded by other studies of bacteria.  It is important to remember that there are differences between prokaryotes and eukaryotes, and these studies encompassed aerobiuc and anaerobic glycolysis, mitochochondrial pathways, and cell death pathways, as well as energy balance, which involves ATP and a series of linked hydrogen transfers in the electron transport chain (ETC).  In the first two papers, we could infer the comparison between differences in oxidative phosphorylation and lactic aciid formation between two strains of cells, whether they be yeast or lymphocytic leukemia.  This is where the observation of Otto Warburg refers back to the work of Pasteur 60 years prior to his discovery.  

In this study we find that metabolic fluxes can be and are measured in Saccharomyces Cerevisiae, and the internal metabolites are measured extensively.  JEDS refers me to Schroedinger’s (Physics Nobel, Quantum Field Theory) classic work, “What is Life?” and his famous CATS, or Rabbits ( a Twilight Zone where objects can be two places at the same time: Schroedinger’s Rabbits: Colin Bruce, Joseph Henry Press, Washington, DC.) That is for another time.  I have also previously referred to the work of Ilya Prigogine (Chemistry Nobel; self organizing systems).  We can set up studies, but we cannot identify the initial state. These two major scientists understood the limits of our ability to study life.

The focus here is on homeostasis.  Homeostasis (Wikipedia), also spelled homoeostasis (from Greek: ὅμοιος, “hómoios”, “similar”,[1] and στάσις, stásis, “standing still”[2]), is the property of a system in which variables are regulated so that internal conditions remain stable and relatively constant. Examples of homeostasis include the regulation of temperature and the balance between acidity and alkalinity (pH). It is a process that maintains the stability of the human body’s internal environment in response to changes in external conditions. The concept was described by Claude Bernard in 1865.  The term was originally used to refer to processes within living organisms, it is frequently applied to automatic control systems.

Buffering of deoxyribonucleotide pool homeostasis by threonine metabolism

John L. Hartman IV *

Author Affiliations   

 Communicated by Leland H. Hartwell, Fred Hutchinson Cancer Research Center, Seattle, WA, June 4, 2007 (received for review January 21, 2007)

Synergistically interacting gene mutations reveal buffering relationships that provide growth homeostasis through their compensation of one another.

Abstract

This analysis in Saccharomyces cerevisiae revealed genetic modules involved in

  • tricarboxylic acid cycle regulation (RTG1RTG2RTG3),
  • threonine biosynthesis (HOM3HOM2HOM6THR1,THR4),
  • amino acid permease trafficking (LST4LST7), and
  • threonine catabolism (GLY1).

These modules contribute to a molecular circuit that

  • regulates threonine metabolism and
  • buffers deficiency in deoxyribonucleotide biosynthesis.

Phenotypic, genetic, and biochemical evidence for this buffering circuit was obtained

  • through analysis of deletion mutants,
  • titratable alleles of ribonucleotide reductase genes, and
  • measurements of intracellular deoxyribonucleotide pool concentrations.

This circuit provides experimental evidence, in eukaryotes, for the presence of a

  • high-flux backbone of metabolism,

which was previously predicted from 

  • in silicomodeling of global metabolism in bacteria.

This part of the high-flux backbone appears to

  • buffer deficiency in ribonucleotide reductase
  • by enabling a compensatory increase in
  • de novopurine biosynthesis
  • that provides additional rate-limiting substrates for
  • dNTP production and DNA synthesis.

Hypotheses regarding unexpected connections

  • between these metabolic pathways
  • were facilitated by genome-wide, and
  • quantitative phenotypic assessment of interactions.

Validation of these hypotheses substantiates

  • the added benefit of quantitative phenotyping
  • for identifying subtleties in gene interactionnetworks
  • that modulate cellular phenotypes.

Keywords: genetic buffering, high-flux backbone of metabolism, protein trafficking, ribonucleotide reductase, mitochondria-to-nucleus retrograde signaling pathway

Introduction 

Cells are complex genetic systems, having evolved

  • compensatory molecular networks
  • that provide growth homeostasis (robustness).

Conceptually, gene interactions

  • underlie robustness by buffering
  • environmental or genetic perturbations (13).

Synergistic effects on the phenotype resulting from

  • two genetic deficiencies or chemical inhibition
  • in combination with a genetic deficiency
  1. reveal buffering relationships
  2. when the double limitation
  3. is more severe than either single limitation.

Genome-wide phenotypic analysis,

  • as possible with RNAi or
  • use of the complete set of
    yeast gene deletion mutants,
  • has enabled new approaches
  • to investigate buffering relationships
    systematically (245).

It has been shown that

  • quantitative (strength) and
  • qualitative (pattern) aspects of
    gene interaction profiles reveal
    • how genes organize
    • in a pathway or cellular process (46).

such sets of genes represent genetic modules that

  • contribute buffering capacity to the cell,
    • providing insight into
    • how molecular circuitry

is arranged to achieve robustness (78).

Comprehensive and quantitative methods

  • for genotype–phenotype analysis are available
    • to gain a more global and precise understanding
    • of buffering networks (469).

These methods permit unbiased

  • investigation of growth homeostasis,
  • systematically revealing
    • how combinations of genetic and
    • environmental variables

result in phenotypic complexity.

High-throughput genotype–phenotype data

  • offer an opportunity to use
  • the extensive and growing genome annotations

to discover new connections between

  • previously annotated genes and pathways,
  • with respect to physiological homeostasis.

Systematic, experimentally derived understanding of

  • genetic interaction networks

will advance efforts to

  • map natural phenotypic variation,
  • thereby aiding the dissection

of genetic disease complexity (10).

This work tests a model constructed after

  • finding threonine biosynthesis
  • to play a role in
  • buffering growth inhibition with

the deoxyribonucleotide (dNTP) biosynthesis
inhibitor, hydroxyurea (HU) (4).

HU is a chemotherapy agent that

  • limits cell proliferation by inhibition of
    ribonucleotide reductase (RNR)
    • leading to dNTP pool deficiency and
    • slow DNA synthesis (11).

The results provide

  • genetic,
  • biochemical, and
  • phenotypic evidence

that growth homeostasis

  • is maintained by
  • a molecular circuit
    • that regulates threonine metabolism
    • to buffer depletion of dNTP pools.

These findings shed light on

  • systems-level observations about
  • cellular metabolism, including
    • function of a high-flux backbone of metabolism (12)
    • and gating of DNA synthesis by
      • oscillation of global transcription
      • and redox metabolism (1314).

FUNCTIONAL INTERACTIONS BETWEEN DNTP AND THREONINE METABOLISM.

This work focused on understanding genetic modules

  • found to buffer RNR deficiency (4).

Synergistic interactions between HU and

  • threonine biosynthesis genes, were uncovered ( 1a).
  • but not genes that function in the synthesis of other amino acids

Deletion ofAAT2 (aspartate aminotransferase) was also synergistic,

  • suggesting buffering by tricarboxylic acid (TCA) cycle flux
  • as AAT2converts the TCA cycle intermediate,
    • oxaloacetate è aspartate
    • the substrate for synthesis of homoserine
    • and ultimately threonine (yeastgenome.org). 

RTG1RTG2, and RTG3, transcription factors

  • regulating transcription of TCA cycle genes
  • in response to mitochondrial stress
    • were also synergistic with hydroxyurea growth limitation
  • further implicating TCA cycle involvement ( 1b).

The RTG and threonine biosynthesis modules were

  • independently confirmed to buffer HU-induced stress
    by Panet al. (17).

Synergistic interaction between HU and

  • deletion alleles ofLST4 and LST7 
  • implicated extracellular uptake of threonine
  • as an alternative mechanism
  • to augment threonine flux ( 1cand ​and22a)
    • becauseLST4 and LST7 regulate
    • delivery of amino acid permeases
      • between the vacuole and
      • plasma membrane compartments of the cell (18).

To confirm that chemical–genetic

  • interactions with HU
  • were caused by its known inhibitory effect
  • on dNTP biosynthesis,
    • a more specific method was used.

Integrating plasmids were

  • introduced into mutant strains
    • to placeRNR1 or RNR2 
  • under transcriptional control by doxycycline (19).

Deletion of homoserine or threonine biosynthesis genes

  • was found to be synergistic with
  • repression of RNR activity by
  • using doxycycline in these mutants ( 1d),
    • confirming that interactions with HU
    • were caused by its inhibitory effect on RNR.

 Media supplementation with amino acids

  • tested whether uptake of extracellular threonine
    • suppresses the growth limitation of mutations
  • in threonine biosynthesis in the presence of HU.

Threonine was found to selectively suppress

  • interaction between HU and
  • disruption of threonine biosynthesis,
  • in a concentration-dependent manner ( 2b–d).

This finding led to the prediction that

  • disabling both threonine biosynthesis and
  • threonine uptake
    • would be synergistic
  • in the presence of HU growth limitation.

 Double deletion mutants of

  • the four possible combinations of
  • thr1or thr4 and lst4 or lst7 were created.

 All combinations were synthetic lethal

  • even in the absence of HU

This is consistent with the hypothesis that lst4 and lst7 

  • compensate threonine biosynthetic deficiency
  • through regulation of extracellular uptake ( 3).

A slow-growth phenotype observed

  • for thehom6 deletion mutant
    • was exacerbated by extracellular threonine,
  • even in the absence of HU (Fig. 2b).

 The hom6deletion mutant is unique

  • among threonine biosynthesis mutants
  • in that the resulting intermediate metabolite is toxic (20)
    (aspartate β-semialdehyde)

Whether this toxicity could be related to its different phenotype

  • in the context of HU perturbation is unexplained.

BIOSYNTHESIS AND EXTRACELLULAR UPTAKE OF THREONINE CONTRIBUTE TO DNTP POOL HOMEOSTASIS.

To test the effect of threonine metabolism on dNTP pools

  • pools were measured in threonine metabolism deletion mutants
  • perturbed by doxycycline-conditional repression
    • of RNR2transcription ( 4).

Although rnr2 deletion is lethal in a haploid,

  • repression of RNR2transcription
  • only reduced the growth rate

when an otherwise non-growth-inhibitory concentration (5 mM) of HU was present (data not shown).

In contrast, RNR1repression was

  • growth-limiting without HU (see  1d) and
  • did not sensitize growth to 5 mM HU
    (data not shown).

The specificity of low-dose HU for

  • growth inhibition in combination with
  • repression of RNR2
  • is explained by the mechanism of action of HU. 

HU scavenges a tyrosyl radical

  • that is present on the Rnr2p and
  • that is required as a cofactor
  • for ribonucleotide reduction (21).

 Non-growth-inhibitory concentrations of HU

  • paradoxically induced increased steady-state
  • dNTP pool concentrations.

The increase in pools was sustained over time

  • and additive with the effect of modulating RNR
    transcriptional levels ( 4a).
  • As a result, growth inhibition,
  • caused byRNR2 repression
  • combined with low-dose HU treatment,
  • occurred with dNTP levels slightly higher than
  • those in the untreated wild-type (WT) control strain
    (endogenousRNR2 promoter).

A possible explanation is that increases in dNTP pools

  • are required for growth fitness
  • in the setting of DNA damage, which is
  • known to involve RNR regulation (22).

 Fig. 1.

HU chemical–genetic interactions. Interactions for three genetic modules are depicted.

HU chemical–genetic interactions. Interactions for three genetic modules are depicted.

HU chemical–genetic interactions. Interactions for three genetic modules are depicted.
The WT control is compared with deletion strains representative of each module,
with respect to their area under the growth curve (AUGC) vs. perturbing drug …

Fig. 2.

Extracellular threonine suppresses interactions between HU and threonine biosynthesis zpq0290770000002

Extracellular threonine suppresses interactions between HU and threonine biosynthesis.
(a) A model explaining interactions between HU and genes involved in threonine metabolism.
In the context of dNTP pool deficiency, threonine metabolism is up-regulated …

To confirm that chemical–genetic interactions with HU

  • were caused by its known inhibitory effect on dNTP biosynthesis,
  • a more specific method was used.

Integrating plasmids were introduced

  • into a variety of mutant strains to place 
  • RNR1or RNR2 under transcriptional control
    by doxycycline (19).

Deletion of homoserine or threonine biosynthesis

  • genes was found to be synergistic with
  • repression of RNR activity by using doxycycline
  • in these mutants ( 1d), confirming that
    • interactions with HU were caused by
      its inhibitory effect on RNR.

Media supplementation with amino acids was used

  • to test whether uptake of extracellular threonine
  • suppresses the growth limitation of mutations
  • in threonine biosynthesis in the presence of HU.

Threonine selectively

  • suppresses interaction between HU and
  • disruption of threonine biosynthesis,
    • in a concentration-dependent manner
      ( 2b–d).

This finding led to the prediction that disabling

  • both threonine biosynthesis and threonine uptake
  • would be synergistic with HU growth limitation.

To test this hypothesis, double deletion mutants of

  • the four possible combinations of
    • thr1or thr4 and lst4 or lst7 were created.

All combinations were synthetic lethal

  • even in the absence of HU,
  • consistent with the hypothesis that
    • lst4andlst7 compensate threonine
      biosynthetic deficiency

through regulation of extracellular uptake (Fig. 3).

A slow-growth phenotype observed for

  • the hom6deletion mutant was exacerbated
  • by extracellular threonine, even
  • in the absence of HU ( 2b).

The hom6deletion mutant is unique among

  • threonine biosynthesis mutants in that
  • the resulting intermediate metabolite is toxic (20)
    (aspartate β-semialdehyde), although
    • whether this toxicity could be related
      to its different phenotype
  • in the context of HU perturbation is unexplained.

Fig. 3.

Deficiency in amino acid permease trafficking (LST4 or LST7) is synthetic lethal with deletion of threonine biosynthesis (THR1 or THR4).

Deficiency in amino acid permease trafficking (LST4 or LST7) is synthetic lethal with deletion of threonine biosynthesis (THR1 or THR4).

Deficiency in amino acid permease trafficking (LST4 or LST7) is synthetic lethal
with deletion of threonine biosynthesis (THR1 or THR4). Interactions between
permease trafficking (LST4 and LST7) and threonine biosynthesis (THR1 and THR4)
were assessed …

BIOSYNTHESIS AND EXTRACELLULAR UPTAKE OF
THREONINE CONTRIBUTE TO DNTP POOL HOMEOSTASIS
.

To test the effect of threonine metabolism on dNTP pools,

  • pools were measured in threonine metabolism deletion mutants
  • perturbed by doxycycline-conditional repression of RNR2transcription ( 4).

Although rnr2 deletion is lethal in a haploid,

  • repression of RNR2transcription only
  • reduced the growth rate when an otherwise
    • non-growth-inhibitory concentration (5 mM)
  • of HU was present (data not shown).

In contrast, RNR1repression was

  • growth-limiting without HU (see  1d)
    • and did not sensitize growth
    • to 5 mM HU (data not shown).

The specificity of low-dose HU for growth inhibition

  • in combination with repression of RNR2
  • is explained by the mechanism of action of HU.

HU scavenges a tyrosyl radical

  • present on the Rnr2p
  • that is required as a cofactor
    • for ribonucleotide reduction (21).

Fig. 4.

Effect of threonine metabolism on dNTP pool homeostasis

Effect of threonine metabolism on dNTP pool homeostasis

Effect of threonine metabolism on dNTP pool homeostasis.
(a) Intracellular dNTP pool concentrations are depicted 90 (black)
and 360 (gray) min after exposure to the perturbations  indicated
by each block. Block 1 is the unperturbed WT (BY4741) strain in …

Non-growth-inhibitory concentrations of HU

  • paradoxically induced increased steady-state
    dNTP pool concentrations.

The increase in pools was sustained over time

  • and additive with the effect of modulating
    RNR transcriptional levels ( 4a).

As a result, growth inhibition, caused by RNR2 repression

  • combined with low-dose HU treatment, occurred
    • with dNTP levels slightly higher than those
    • in the untreated wild-type (WT) control strain
      (endogenous RNR2promoter).

A possible explanation is that increases in dNTP pools are

  • required for growth fitness in the setting of DNA damage,
  • which is known to involve RNR regulation (22).

However, production of DNA damage
(requiring increased dNTP pools for DNA repair)

  • would have been expected only at HU concentrations
  • high enough to arrest DNA synthesis in the first place (23).

The observation that low concentrations of HU

  • led to increased pools could be explained
  • if DNA damage occurs by a mechanism
    • independent of the effect of HU
    • on cytoplasmic pools.

A possible mechanism could involve

  • dNTP pool concentrations at replication forks
  • being affected differentially from
    • cytoplasmic pools;
  • this is not thought to occur
    in eukaryotic cells (24).

Thus, the paradoxical effect of

  • low HU concentrations
  • on increasing dNTP pools
  • remains unexplained.

dNTP pools were increased

  • by expression ofRNR2  from the Tet promoter
    (in the absence of repression with doxycycline),
  • presumably because of overexpression
  • relative to the endogenousRNR2 

Dox-conditional repression of RNR2 

  • reduced dNTP pools in a
  • concentration-dependent fashion ( 4a)
    • so that synergism from deletion of
      threonine metabolism genes could be tested.

The rtg2hom2thr1, and lst4 deletion mutants

  • all exacerbated the reduction in dNTP pools
    • afterRNR2 repression ( 4b).

The contribution of RTG2 for

  • dNTP pool maintenance
  • was less than that of HOM2,THR1, or LST4,
  • consistent with their effects on growth ( 1).

 Consistent with low dNTP pools

  • causing cell cycle arrest in each of the mutants,
    • median cell size increased
    • the relative number of cells and
    • total cell volume
      (median cell size × median cell volume)
    • decreased as pools became depleted ( 4c).

The scs7 (functions in sphingolipid metabolism) deletion strain

  • maintained dNTP pools comparable with WT,
  • despite a greater fitness defect ( 4b and c),
    • indicating a specific role of threonine metabolic genes
    • in homeostatic regulation of dNTP pools.

THREONINE ALDOLASE IS RATE-LIMITING FOR DNTP METABOLISM IN SACCHAROMYCES CEREVISIAE.

The genetic, phenotypic, and biochemical results

  • are consistent with a model whereby
    • TCA cycle regulation (RTG genes),
    • threonine biosynthesis (HOM and THR genes), and
    • permease trafficking (LST genes) pathways
    • coordinately buffer dNTP pool depletion by
      • up-regulating threonine metabolism.

The model postulates that threonine catabolism

  • contributes glycine to augment
  • de novopurine synthesis ( 2a).

HU has been shown to preferentially deplete

  • dATP pools in mammalian cells (2526), and
  • there was a tendency for purine pools to fluctuate
    (particularly dATP)
    • acutely whenever threonine metabolism and
    • RNR activity were perturbed in combination
      ( 4aand b).

However, allosteric regulation of RNR would

  • distribute this effect across all pools (21).

Threonine aldolase, encoded by GLY1 (EC 4.1.2.5),

  • cleaves threonine into glycine and acetaldehyde (27).

Notably, the gly1 deletion mutant exhibited slow growth
(data not shown)

  • even with glycine supplementation.

This phenotype was found to be the result of

  • limitation of dNTP metabolism.

Basal dNTP pools were reduced

  • in the gly1deletion mutant,

Pools fell dramatically

  • after treatment with 10 mM HU,

and normal homeostatic increases in dNTP concentrations

  • after treatment with 50 mM HU were delayed,
  • particularly dATP pools (Fig. 5). 

CHA1 (EC 4.2.1.13) and ILV1 (EC 4.3.1.19) are

  • deaminases that convert threonine to 2-oxybutanoate
  • or other metabolic intermediates such as
    • homoserine, cystathionine, or propionyl-CoA.

However, deletion of neither CHA1 nor ILV1 

  • modified the growth response to HU (4).

Fig. 5.

Threonine aldolase contributes to normal dNTP metabolism

Threonine aldolase contributes to normal dNTP metabolism

Threonine aldolase contributes to normal dNTP metabolism. Intracellular dNTP pools are shown for the WT control strain (BY4741) and gly1 (threonine aldolase) deletion mutant before (Left) and 120 or 360 min after perturbation with 10 mM (Center) or 50 

DISCUSSION

Computational analysis of global metabolism

  • inEscherichia coli has suggested
  • that threonine flux is of particular importance.

These studies propose that

  • threonine synthesis and
  • its degradation to glycine
    • for purine biosynthesis
  • are part of a high-flux backbone (HFB)
  • of metabolism (12).

The HFB was defined by a

  • subset of all metabolic reactions
  • found to have sufficient flux for
    • providing growth homeostasis
  • in response to growth-limiting perturbations
    (such shifting to a poor carbon source).

Utilization of threonine for buffering

  • dNTP metabolism and growth homeostasis
  • provides experimental evidence for
  • the presence of the HFB in eukaryotes.

Discovery of new connections between

  • dNTP and threonine metabolism
  1. demonstrates the value of quantitative high-throughput
    cellular phenotyping for identifying 
  2. functional redundancies in gene networks
    • by measuring interactions between
    • genetic module metabolism.

The ability to detect relatively small effects

  • of individual modules and
  • to order their relative quantitative impact
    • aided hypotheses about how
    • these modules might relate to one another (4).

By this approach, genes involved in

  1. TCA cycle regulation,
  2. threonine biosynthesis,
  3. amino acid permease trafficking,
  4. threonine catabolism, and
  5. ribonucleotide reduction
  • were found to function as a modular circuit
  • to maintain robust dNTP pools
  • for DNA synthesis
    • even though these modules
    • appear to function independently
      in other contexts (1835).

In natural (outbred) populations,

  • compensatory networks also buffer
  • genetic and chemical growth perturbations;

however, the amount of genotypic and phenotypic variation

  • renders dissection of interactions relatively intractable.

By contrast, systematic analysis of yeast deletion mutants

  • exposes interactions on a fixed genetic background
    • but does not survey natural variation.

Recently, segregants from a cross of S288C
(the background used for systematic gene deletion)

  • and a natural isolate
  • have been genotyped at high resolution (3637).

Quantitative high-throughput cellular phenotyping,

  • applied in parallel to these strains and
  • the comprehensive collection of
  • yeast gene deletion mutants, would
    • provide a dual strategy to
    • deconstruct gene networks
    • that buffer growth perturbations, by
    • systematic analysis of all deletion mutants
    • in parallel with surveying for natural occurrence.

Quantitative genetic dissection of

  • buffering networks in yeast thus
  • provides a way to model genotype–phenotype variation
  • on a genomic scale, providing insight into
    • functional interactions between conserved pathways
    • that potentially modulate human disease.

Cell Proliferation Measurements. 

Experiments represented in Figs. 1 and ​and22 were performed in Hartwell complete agar medium. High-throughput kinetic phenotyping (by imaging and image analysis) and area under the growth curve (AUGC) calculations were performed as described previously (4). AUGC encapsulates the overall growth phenotype of a strain with respect to time under a particular condition. AUGC is affected by initial population size (no. of cells transferred in a spot culture), lag time (delay before log-linear growth), maximum specific rate (actual log-linear rate), total efficiency (saturation density), and duration of the assay. For assessing the strength of a genetic interaction, the change in the AUGC conferred by a particular deletion allele relative to its WT control allele is considered with respect to perturbation intensity, e.g., concentration of HU, as depicted in Fig. 1. AUGC values for all mutants perturbed with 0, 50, and 150 mM HU are available at (http://genomebiology.com/2004/5/7/R49/additional).

dNTP Pool Sample Collections. 

Strains were grown overnight in liquid medium at 30°C to a concentration of ≈3 × 106 cells/ml and diluted to prewarmed medium with HU or doxycycline to achieve the desired cell and drug concentrations in a final volume of 30 ml. Each time point was grown separately and harvested when the cell concentration was ≈3 × 106 cells per ml. Twenty milliliters of culture was collected by vacuum filtration and immediately washed with ice-cold medium, and filters were transferred to 2 ml of ice-cold medium (dNTP concentrations remain stable in iced medium for several hours). Cells were removed from the filter by vortexing, the sample was divided in half for duplicate readings, cells were pelleted, medium removed by aspiration, and cells were lysed with 40 μl of 0.1 M perchloric acid and then snap-frozen.

Cell Volume Measurements. 

Cell volumes were measured by size analysis with a Coulter Counter (Beckman–Coulter, Fullerton, CA). The total cell volume of each culture (median cell size × total cell number) was used for calculating intracellular dNTP pool concentrations. Before vacuum filtration and lysis of each culture for mass spectrometry analysis, 200 μl was collected into 10 ml of ice-cold isoton (Beckman–Coulter). Samples were sonicated at low power to separate nonspecifically adherent cells. To calculate relative changes in total cell volume (Fig. 4c), values for each strain were first normalized against self at time zero and then divided by the corresponding normalized WT (BY4741) values.

HPLC. 

Samples were thawed by microcentrifugation (18,000 × g) for 15 min at 4°C. Sixteen microliters of lysate was added to 8 μl of 3× mobile-phase buffer [60 mM acetic acid/0.075% dimethylhydroxylamine (Sigma, St. Louis, MO)/pH adjusted to 7 with ammonium hydroxide], and 10 μl was injected onto an Agilent C-8 Zorbax column (part 883700-906) with a linear 5–30% methanol gradient from 2 to 11 min, 30–50% from 11 to 12 min, with final reequilibration for 5 min in 5% methanol (flow rate of 0.3 ml/min). Retention times of 4.5 (dTTP), 7.5 (dGTP and dTTP), and 9.5 min (dATP) were observed. dNTP-depleted lysate was obtained by lysis of saturation-density cultures after 30-min incubation in room temperature water. Dilution of standards in this lysate improved dCTP chromatography. Trace amounts of dNTPs remaining in the diluent were subtracted for standard curve calculations.

Mass Spectrometry. 

Mass spectrometry was performed with electron spray ionization in negative ion mode. Two instruments were used: (i) an Agilent 1100 MSD [dNTPs were monitored as single ions at m/z 466 (dCTP)], 481 (dTTP), 490 (dTTP), and 506 (dGTP). The drying gas was N2 at 340°C at 10 liters/min, and nebulizing pressure 25 psi (1 psi = 6.89 kPa). The fragmentor was set at 90 eV and capillary voltage 3500. (ii) An ABI API-4000 Q-trap triple quadrupole instrument was used [mass transition to a 189 fragment was monitored for each of the dNTP species, as described previously (39); N2 gas was used for nebulization, drying, and collision and the ionization chamber temperature was 250°C]. New standard curves were created for every assay.

Calculation of Intracellular dNTP Concentrations. 

Sample concentrations were determined from standard curves and adjusted to account for dilution by lysis and total cell volume [volume added for lysis + 2(tcv)] μL /tcv (μL). Standard curves showed high linear correlation (R2 > 0.998), and variation from duplicate mass spec measurements was generally <10%.

ABBREVIATIONS: AUGC, area under the growth curve; dNTP, deoxyribonucleotide; HU, hydroxyurea; RNR, ribonucleotide reductase; TCA, tricarboxylic acid..

The requirement reported here of mitochondrial-to-nucleus retrograde signaling for dNTP pool homoeostasis in yeast may be of importance to a recent report that mutations in p53R2 cause human mitochondrial depletion syndromes (MDS) (4041). If compensatory/buffering relationships between RNR and retrograde signaling in yeast are evolutionarily conserved, then genetic variation in retrograde signaling may modulate MDS disease phenotypes resulting from deficiency in p53R2 activity.

ARTICLE INFORMATION

Proc Natl Acad Sci U S A. Jul 10, 2007; 104(28): 11700–11705.

Published online Jul 2, 2007. doi:  10.1073/pnas.0705212104

PMCID: PMC1913885

Genetics

John L. Hartman, IV*

Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294

*To whom correspondence should be addressed. E-mail: ude.bau@namtrahj

Communicated by Leland H. Hartwell, Fred Hutchinson Cancer Research Center, Seattle, WA, June 4, 2007.

Author contributions: J.L.H. designed research, performed research, contributed new reagents/analytic tools,
analyzed data, and wrote the paper.

Received January 21, 2007

Buffering of deoxyribonucleotide pool homeostasis by threonine metabolism

John L. Hartman, IV

Additional article information

NOTE ADDED IN PROOF.

Note Added in Proof.

The requirement reported here of mitochondrial-to-nucleus retrograde signaling for dNTP pool homoeostasis in yeast may be of importance
to a recent report that mutations in p53R2 cause human mitochondrial depletion syndromes (MDS) (4041). If compensatory/buffering
relationships between RNR and retrograde signaling in yeast are evolutionarily conserved, then genetic variation in retrograde signaling
may modulate MDS disease phenotypes resulting from deficiency in p53R2 activity.

FOOTNOTES

The author declares no conflict of interest.

ARTICLE INFORMATION

Proc Natl Acad Sci U S A. Jul 10, 2007; 104(28): 11700–11705.

Published online Jul 2, 2007. http://dx.doi.org:/10.1073/pnas.0705212104

PMCID: PMC1913885

Genetics

John L. Hartman, IV*

Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294

*To whom correspondence should be addressed. E-mail: ude.bau@namtrahj

Communicated by Leland H. Hartwell, Fred Hutchinson Cancer Research Center, Seattle, WA, June 4, 2007.

Author contributions: J.L.H. designed research, performed research, contributed new reagents/analytic tools, analyzed data, and wrote the paper.

Received January 21, 2007  Copyright © 2007 by The National Academy of Sciences of the USA

This article has been cited by other articles in PMC.

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

 REFERENCES

  1. Hartman JL, IV, Garvik B, Hartwell L. Science. 2001;291:1001–1004. [PubMed]
  2. Tong AH, Lesage G, Bader GD, Ding H, Xu H, Xin X, Young J, Berriz GF, Brost RL, Chang M, et al. Science. 2004;303:808–813. [PubMed]
  3. Lehner B, Crombie C, Tischler J, Fortunato A, Fraser AG. Nat Genet. 2006;38:896–903.[PubMed]
  4. Hartman JL, IV, Tippery NP. Genome Biol. 2004;5:R49. [PMC free article] [PubMed]
  5. Parsons AB, Brost RL, Ding H, Li Z, Zhang C, Sheikh B, Brown GW, Kane PM, Hughes TR, Boone C. Nat Biotechnol. 2004;22:62–69. [PubMed]
  6. Collins SR, Schuldiner M, Krogan NJ, Weissman JS. Genome Biol. 2006;7:R63.[PMC free article] [PubMed]
  7. Csete ME, Doyle JC. Science. 2002;295:1664–1669. [PubMed]
  8. Hartwell LH, Hopfield JJ, Leibler S, Murray AW. Nature. 1999;402:C47–C52. [PubMed]
  9. Shah NA, Laws RJ, Wardman B, Zhao LP, Hartman JL., IV BMC Syst Biol. 2007;1:3.[PMC free article] [PubMed]
  10. Badano JL, Katsanis N. Nat Rev Genet. 2002;3:779–789. [PubMed]
  11. Krakoff IH, Brown NC, Reichard P. Cancer Res. 1968;28:1559–1565. [PubMed]
  12. Almaas E, Kovacs B, Vicsek T, Oltvai ZN, Barabasi AL. Nature. 2004;427:839–843.[PubMed]

 …..

Read Full Post »

Cell Research News – What’s to Follow?

Larry H. Bernstein, MD, FCAP, Reporter

Leaders in Pharmaceutical Intelligence

http://pharmaceuticalintelligence.com/2014/08/26/larryhbern/Cell_Research_News_-_What’s_to_Follow?

 

Stem Cell Research ‘Holy Grail’ Uncovered, Thanks to Zebrafish

By Estel Grace Masangkay

With help from the zebrafish, a team of Australian researchers has uncovered how
hematopoietic stem cells (HSC) renew themselves.

HSCs refers to stem cells present in the blood and bone marrow that are used 
for  the replenishment of the body’s supply of blood and immune cells – 

  • in transplants for leukemia and myeloma.
  • Stem cells have the potential to transform into vital cells

    including muscle, bone, and blood vessels.

Understanding how HSCs form and renew themselves has potential application in the
treatment of

  • spinal cord injuries
  • degenerative disorders
  • diabetes.

Professor Peter Currie, of the Australian Regen Med Institute at Victoria’s Monash
University, led a research team to discover a crucial part of HSC’s development. Using 
a high-resolution microscopy, Prof. Curie’s team 

  • caught zebrafish embyonic SCs on film as they formed. 
  • the researchers were studying muscle mutations in the aquatic animal.

“Zebrafish make ESCs in exactly the same way as humans do, but their embryos and
larvae develop free living, but the larvae are both free swimming and transparent, so one could see every cell in the body forming, including ESCs,” explained Prof. Currie.

The researchers noticed in films that a

  •  ‘buddy cell’ came along to help the ESCs form.

Called endotome cells, 

  • they aided pre-ESCs to turn into ESCs.  

Prof. Currie said that endotome cells act as helper cells for pre-ESCs , 

  • helping them progress to become fully fledged stem cells.

The team not only

  • identified some of the cells and signals 
  • required for ESC formation, but also 
  • pinpointed the genes required 
  • for endotome formation in the first place.

The next step for the researchers is to 

  • locate the signals present in the endotome cells 
  • that trigger ESC formation in the embryo. 

This may provide clues for developing

  • specific blood cells on demand for blood-related disorders. 

Professor Currie also pointed out the discovery’s potential for 

  • correcting genetic defects in the cell and 
  • transplanting them back in the body to treat disorders.

The team’s work was published in the international journal Nature.

 

Jell-O Like Biomaterial Could Hold Key to Cancer Cell Destruction

by Estel Grace Masangkay

Scientists from Penn State University reported that a biomaterial made of tiny 
molecules was able to attract and destroy cancer cells.

Professor Yong Wang and bioengineering faculty at Penn State, built the 
tissue-like biomaterial to accomplish what chemotherapy could not –

  • kill every cancer cell without leaving
  • the possibility of a recurrence.

Prof. Wang and team built polymers 

  • from tiny molecules called monomers. They
  • then wove the polymers into 3D networks 

called hydrogels. Hydrogel is soft and flexible, 
like Jell-O, and it contains a lot of water, and

  • can be safely put into the body, unlike 

other implants that the body often tries 

  • to get rid of through the immune response.

“We want to make sure the materials we are using are compatible in the body.”

The researchers 

  • attached aptamers to the hydrogels, 
  • which release bio-chemical signal-only molecules 
  • that draw in cancer cells. 

Once attracted, the cancer cells are entrapped in the Jell-O-like substance. 

What happens next is 

  • an oligonucleotide binds to the protein-binding site of the aptamer 
  • and triggers the release of anticancer drugs at the proper time.

“Once we trap the cancer cells, we can deliver anticancer drugs 

  • to that specific location to kill them. 

This technique would help avoid the need for systemic medications that kill not only cancer cells, but normal cells as well. Systemic chemotherapy drugs

  • make patients devastatingly sick and possibly 
  • leave behind cancer cells to wreak havoc another day

If our new technique has any side effects at all, it would be only local side 
effects and not whole-body systemic side effects,” explained Prof. Wang.

The initial results of the research were published by Prof. Wang in the 
Journal of the American Chemical Society in 2012. Prof. Wang also shared 
the latest results of his work at the Society for Biomaterials Meeting &
 Exposition in April this year.

 

Read Full Post »

Extracellular evaluation of intracellular flux in yeast cells

Larry H. Bernstein, MD, FCAP, Reviewer and Curator

Leaders in Pharmaceutical Intelligence

This is the fourth article in a series on metabolomics, which is a major development in -omics, integrating transcriptomics, proteomics,  genomics, metabolic pathways analysis, metabolic and genomic regulatory control using computational mapping.  In the previous two part presentation, flux analysis was not a topic for evaluation, but here it is the major focus.  It is a study of yeast cells, and bears some relationship to the comparison of glycemia, oxidative phosphorylation, TCA cycle, and ETC in leukemia cell lines.  In the previous study – system flux was beyond the scope of analysis, and explicitly stated.  The inferences made in comparing the two lymphocytic leukemia cells was of intracellular metabolism from extracellular measurements.  The study of yeast cells is aimed at looking at cellular effluxes, which is also an important method for studying pharmacological effects and drug resistance.

Metabolomic series

1.  Metabolomics, Metabonomics and Functional Nutrition: the next step in nutritional metabolism and biotherapeutics

http://pharmaceuticalintelligence.com/2014/08/22/metabolomics-metabonomics-and-functional-nutrition-the-next-step-in-nutritional-metabolism-and-biotherapeutics/

2.  Metabolomic analysis of two leukemia cell lines. I

http://pharmaceuticalintelligence.com/2014/08/23/metabolomic-analysis-of-two-leukemia-cell-lines-_i/

3.  Metabolomic analysis of two leukemia cell lines. II.

 http://pharmaceuticalintelligence.com/2014/08/24/metabolomic-analysis-of-two-leukemia-cell-lines-ii/

4.  Extracellular evaluation of intracellular flux in yeast cells

Q1. What is efflux?

Q2. What measurements were excluded from the previous study that would not allow inference about fluxes?

Q3. Would this study bear any relationship to the Pasteur effect?

Q4 What is a genome scale network reconstruction?

Q5 What type of information is required for a network prediction model?

Q6. Is there a difference between the metabolites profiles for yeast grown under aerobic and anaerobuc conditions – under the constrainsts?

Q7.  If there is a difference in the S metabolism, would there be an effect on ATP production?

 

 

Connecting extracellular metabolomic measurements to intracellular flux
states in yeast

Monica L Mo1Bernhard Ø Palsson1 and Markus J Herrgård12*

Author Affiliations

1 Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA

2 Current address: Synthetic Genomics, Inc, 11149 N Torrey Pines Rd, La Jolla, CA 92037, USA

For all author emails, please log on.

BMC Systems Biology 2009, 3:37  doi:10.1186/1752-0509-3-37

 

The electronic version of this article is the complete one and can be found online at: http://www.biomedcentral.com/1752-0509/3/37

 

Received: 15 December 2008
Accepted: 25 March 2009
Published: 25 March 2009

© 2009 Mo et al; licensee BioMed Central Ltd.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Background

Metabolomics has emerged as a powerful tool in the

  • quantitative identification of physiological and disease-induced biological states.

Extracellular metabolome or metabolic profiling data, in particular,

  • can provide an insightful view of intracellular physiological states in a noninvasive manner.

Results

We used an updated genome-scale

  • metabolic network model of Saccharomyces cerevisiae, iMM904, to investigate
  1. how changes in the extracellular metabolome can be used
  2. to study systemic changes in intracellular metabolic states.

The iMM904 metabolic network was reconstructed based on

  • an existing genome-scale network, iND750,
  • and includes 904 genes and 1,412 reactions.

The network model was first validated by

  • comparing 2,888 in silico single-gene deletion strain growth phenotype predictions
  • to published experimental data.

Extracellular metabolome data measured

  • of ammonium assimilation pathways 
  • in response to environmental and genetic perturbations

was then integrated with the iMM904 network

  • in the form of relative overflow secretion constraints and
  • a flux sampling approach was used to characterize candidate flux distributions allowed by these constraints.

Predicted intracellular flux changes were

  • consistent with published measurements
  • on intracellular metabolite levels and fluxes.

Patterns of predicted intracellular flux changes

  • could also be used to correctly identify the regions of
  • the metabolic network that were perturbed.

Conclusion

Our results indicate that

  • integrating quantitative extracellular metabolomic profiles
  • in a constraint-based framework
  • enables inferring changes in intracellular metabolic flux states.

Similar methods could potentially be applied

  • towards analyzing biofluid metabolome variations
  • related to human physiological and disease states.

Background

“Omics” technologies are rapidly generating high amounts of data

  • at varying levels of biological detail.

In addition, there is a rapidly growing literature and

  • accompanying databases that compile this information.

This has provided the basis for the assembly of

  • genome-scale metabolic networks for various microbial and eukaryotic organisms [111].

These network reconstructions serve

  • as manually curated knowledge bases of
  • biological information as well as
  • mathematical representations of biochemical components and
  • interactions specific to each organism.

genome-scale network reconstruction is

  • structured collection of genes, proteins, biochemical reactions, and metabolites
  • determined to exist and operate within a particular organism.

This network can be converted into a predictive model

  • that enables in silico simulations of allowable network states based on
  • governing physico-chemical and genetic constraints [12,13].

A wide range of constraint-based methods have been developed and applied

  • to analyze network metabolic capabilities under
  • different environmental and genetic conditions [13].

These methods have been extensively used to

  • study genome-scale metabolic networks and have successfully predicted, for example,
  1. optimal metabolic states,
  2. gene deletion lethality, and
  3. adaptive evolutionary endpoints [1416].

Most of these applications utilize

  • optimization-based methods such as flux balance analysis (FBA)
  • to explore the metabolic flux space.

However, the behavior of genome-scale metabolic networks can also be studied

  • using unbiased approaches such as
  • uniform random sampling of steady-state flux distributions [17].

Instead of identifying a single optimal flux distribution based on

  • a given optimization criterion (e.g. biomass production),

these methods allow statistical analysis of

  • a large range of possible alternative flux solutions determined by
  • constraints imposed on the network.

Sampling methods have been previously used to study

  1. global organization of E. coli metabolism [18] as well as
  2. to identify candidate disease states in the cardiomyocyte mitochondria [19].

Network reconstructions provide a structured framework

  • to systematically integrate and analyze disparate datasets
  • including transcriptomic, proteomic, metabolomic, and fluxomic data.

Metabolomic data is one of the more relevant data types for this type of analysis as

  1. network reconstructions define the biochemical links between metabolites, and
  2. recent advancements in analytical technologies have allowed increasingly comprehensive
  • intracellular and extracellular metabolite level measurements [20,21].

The metabolome is

  1. the set of metabolites present under a given physiological condition
  2. at a particular time and is the culminating phenotype resulting from
  • various “upstream” control mechanisms of metabolic processes.

Of particular interest to this present study are

  • the quantitative profiles of metabolites that are secreted into the extracellular environment
  • by cells under different conditions.

Recent advances in profiling the extracellular metabolome (EM) have allowed

  • obtaining insightful biological information on cellular metabolism
  • without disrupting the cell itself.

This information can be obtained through various

  • analytical detection,
  • identification, and
  • quantization techniques

for a variety of systems ranging from

  • unicellular model organisms to human biofluids [2023].

Metabolite secretion by a cell reflects its internal metabolic state, and

  • its composition varies in response to
  • genetic or experimental perturbations
  • due to changes in intracellular pathway activities
  • involved in the production and utilization of extracellular metabolites [21].

Variations in metabolic fluxes can be reflected in EM changes which can

  • provide insight into the intracellular pathway activities related to metabolite secretion.

The extracellular metabolomic approach has already shown promise

  • in a variety of applications, including
  1. capturing detailed metabolite biomarker variations related to disease and
  2. drug-induced states and
  3. characterizing gene functions in yeast [2427].

However, interpreting changes in the extracellular metabolome can be challenging

  • due to the indirect relationship between the proximal cause of the change
    (e.g. a mutation)
  • and metabolite secretion.

Since metabolic networks describe

  • mechanistic,
  • biochemical links between metabolites,

integrating such data can allow a systematic approach

  • to identifying altered pathways linked to
  • quantitative changes in secretion profiles.

Measured secretion rates of major byproduct metabolites

  • can be applied as additional exchange flux constraints
  • that define observed metabolic behavior.

For example, a recent study integrating small-scale EM data

  • with a genome-scale yeast model
  • correctly predicted oxygen consumption and ethanol production capacities
  • in mutant strains with respiratory deficiencies [28].

The respiratory deficient mutant study

  • used high accuracy measurements for a small number of
  • major byproduct secretion rates
  • together with an optimization-based method well suited for such data.

Here, we expand the application range of the model-based method used in [28]

  • to extracellular metabolome profiles,
  • which represent a temporal snapshot of the relative abundance
  • for a larger number of secreted metabolites.

Our approach is complementary to

  • statistical (i.e. “top-down”) approaches to metabolome analysis [29]
  • and can potentially be used in applications such as biofluid-based diagnostics or
  • large-scale characterization of mutants strains using metabolite profiles.

This study implements a constraint-based sampling approach on

  • an updated genome-scale network of yeast metabolism
  • to systematically determine how EM level variations

are linked to global changes in intracellular metabolic flux states.

By using a sampling-based network approach and statistical methods (Figure 1),

  • EM changes were linked to systemic intracellular flux perturbations
    in an unbiased manner
  • without relying on defining single optimal flux distributions
  • used in the previously mentioned study [28].

The inferred perturbations in intracellular reaction fluxes were further analyzed

  • using reporter metabolite and subsystem (i.e., metabolic pathway) approaches [30]
  • in order to identify dominant metabolic features that are collectively perturbed (Figure 2).

The sampling-based approach also has the additional benefit of

  • being less sensitive to inaccuracies in metabolite secretion profiles than
  • optimization-based methods and can effectively be used – in biofluid metabolome analysis.
integration of exometabolomic (EM) data

integration of exometabolomic (EM) data

Figure 1. Schematic illustrating the integration of exometabolomic (EM) data with the constraint-based framework.

(A) Cells are subjected to genetic and/or environmental perturbations to secrete metabolite patterns unique to that condition.
(B) EM is detected, identified, and quantified.
(C) EM data is integrated as required secretion flux constraints to define allowable solution space.
(D) Random sampling of solution space yields the range of feasible flux distributions for intracellular reactions.
(E) Sampled fluxes were compared to sampled fluxes of another condition to determine

  • which metabolic regions were altered between the two conditions (see Figure 2).

(F) Significantly altered metabolic regions were identified.

http://www.biomedcentral.com/content/figures/1752-0509-3-37-1.jpg

 

sampling and scoring analysis to determine intracellular flux changes

sampling and scoring analysis to determine intracellular flux changes

Figure 2. Schematic of sampling and scoring analysis to determine intracellular flux changes.

(A) Reaction fluxes are sampled for two conditions.
(B & C) Sample of flux differences is calculated by selecting random flux values from each condition

  • to obtain a distribution of flux differences for each reaction.

(D) Standardized reaction Z-scores are determined, which represent

  • how far the sampled flux differences deviates from a zero flux change.

Reaction scores can be used in

  1. visualizing perturbation subnetworks and
  2. analyzing reporter metabolites and subsystems.

http://www.biomedcentral.com/content/figures/1752-0509-3-37-2.jpg

This study was divided into two parts and describes:

(i) the reconstruction and validation of an expanded S. cerevisiae metabolic network, iMM904; and
(ii) the systematic inference of intracellular metabolic states from

  • two yeast EM data sets using a constraint-based sampling approach.

The first EM data set compares wild type yeast to the gdh1/GDH2 (glutamate dehydrogenase) strain [31],

  • which indicated good agreement between predicted metabolic changes
  • of intracellular metabolite levels and fluxes [31,32].

The second EM data set focused on secreted amino acid measurements

  • from a separate study of yeast cultured in different
    ammonium and potassium concentrations [33].

We analyzed the EM data to gain further insight into

  • perturbed ammonium assimilation processes as well as
  1. metabolic states relating potassium limitation and
  2. ammonium excess conditions to one another.

The model-based analysis of both

  • separately published extracellular metabolome datasets
  • suggests a relationship between
  1. glutamate,
  2. threonine and
  3. folate metabolism,
  • which are collectively perturbed when
    ammonium assimilation processes are broadly disrupted
  1. either by environmental (excess ammonia) or
  2. genetic (gene deletion/overexpression) perturbations.

The methods herein present an approach to

  • interpreting extracellular metabolome data and
  • associating these measured secreted metabolite variations
  • to changes in intracellular metabolic network states.

Additional file 1. iMM904 network content.

The data provided represent the content description of the iMM904 metabolic network and
detailed information on the expanded content.

Format: XLS Size: 2.7MB Download file

This file can be viewed with: Microsoft Excel Viewer

Additional file 2. iMM904 model files.

The data provided are the model text files of the iMM904 metabolic network
that is compatible with the available COBRA Toolbox [13]. The model structure
can be loaded into Matlab using the ‘SimPhenyPlus’ format with GPR and compound information.

Format: ZIP Size: 163KB Download file

Conversion of the network to a predictive model

The network reconstruction was converted to a constraint-based model using established procedures [13].

Network reactions and metabolites were assembled into a stoichiometric matrix 

  • containing the stoichiometric coefficients of the reactions in the network.

The steady-state solution space containing possible flux distributions

  • is determined by calculating the null space of S= 0,

where is the reaction flux vector.

Minimal media conditions were set through constraints on exchange fluxes

  • corresponding to the experimental measured substrate uptake rates.

All the model-based calculations were done using the Matlab COBRA Toolbox [13]

  • utilizing the glpk or Tomlab/CPLEX (Tomopt, Inc.) optimization solvers.

Chemostat growth simulations

The iMM904 model was initially validated by

  1. simulating wild type yeast growth in aerobic and anaerobic
    carbon-limited chemostat conditions
  2. and comparing the simulation results to published experimental data

on substrate uptake and byproduct secretion in these conditions [34].

The study was performed following the approach taken to validate the iFF708 model in a previous study [35].

The predicted glucose uptake rates were determined

  1. by setting the in silico growth rate to the measured dilution rate,
    – equivalent under continuous culture growth,
  2. and minimizing the glucose uptake rate.

The accuracy of in silico predictions of

  • substrate uptake and byproduct secretion by the iMM904 model
  • was similar to the accuracy obtained using the iFF708 model
  • and results are shown in Figure S1 [see Additional file 3].

Additional file 3. Supplemental figures. 

The file provides the supplemental figures and descriptions of S1, S2, S3, and S4.

Format: PDF Size: 513KB Download file

This file can be viewed with: Adobe Acrobat Reader

Genome-scale gene deletion phenotype predictions

The iMM904 network was further validated by

  • performing genome-scale gene lethality computations
  • following established procedures to determine growth phenotypes
  1. under minimal medium conditions and
  2. compared to published data.

A modified version of the biomass function used in previous iND750 studies

  1. was set as the objective to be maximized and
  2. gene deletions were simulated by

setting the flux through the corresponding reaction(s) to zero.

The biomass function was based on the experimentally measured

  1. composition of major cellular constituents
  2. during exponential growth of yeast cells and
  3. was reformulated to include trace amounts of
  4. additional cofactors and metabolites
  5. with the assumed fractional contribution of 10-.

These additional biomass compounds were included

according to the biomass formulation used in the iLL672 study

  • to improve lethality predictions through
  • the inclusion of additional essential biomass components [3].

The model was constrained by limiting

  1. the carbon source uptake to 10 mmol/h/gDW
  2. and oxygen uptake to 2 mmol/h/gDW.

Ammonia, phosphate, and sulfate were assumed to be non-limiting.

The experimental phenotyping data was obtained

  • using strains that were auxotrophic for
  1. methionine,
  2. leucine,
  3. histidine, and
  4. uracil.

These auxotrophies were simulated

  1. by deleting the appropriate genes from the model and
  2. supplementing the in silico strain with the appropriate supplements
  3. at non-limiting, but low levels.

Furthermore, trace amounts of essential nutrients that are present

  • in the experimental minimal media formulation
  1. 4-aminobenzoate,
  2. biotin,
  3. inositol,
  4. nicotinate,
  5. panthothenate,
  6. thiamin)
  • were supplied in the simulations [3].

Three distinct methods to simulate the outcome of gene deletions were utilized:

  1. Flux-balance analysis (FBA) [36-38],
  2. Minimization of Metabolic Adjustment (MoMA) [39], and
  3. a linear version of MoMA (linearMoMA).

In the linearMoMA method, minimization of the quadratic objective function
of the original MoMA algorithm

  • was replaced by minimization of the corresponding 1-norm objective function
    (i.e. sum of the absolute values of the differences of wild type FBA solution
    and the knockout strain flux solution).

The computed results were then compared to growth phenotype data
(viable/lethal) from a previously published experimental gene deletion study [3].

The comparison between experimental and in silico deletion phenotypes involved

  • choosing a threshold for the predicted relative growth rate of
  • a deletion strain that is considered to be viable.

We used standard ROC curve analysis

  • to assess the accuracy of different prediction methods and models
  • across the full range of the viability threshold parameter,
    results shown in Figure S2 [see Additional file 3].

The ROC curve plots the true viable rate against the false viable rate

  • allowing comparison of different models and methods
  • without requiring arbitrarily choosing this parameter a priori [40].

The optimal prediction performance corresponds to

  • the point closest to the top left corner of the ROC plot
    (i.e. 100% true viable rate, 0% false viable rate).

Table 1

Table 1 Comparison of iMM904 and iLL672 gene deletion predictions and experimental data under minimal media conditions
Media Model Method True viable False viable False lethal True lethal True viable % False viable % MCC
Glucose iMM904 full FBA 647 10 32 33 95.29 23.26 0.6
iMM904 full linMOMA 644 10 35 33 94.85 23.26 0.58
iMM904 full MOMA 644 10 35 33 94.85 23.26 0.58
iMM904 red FBA 440 9 28 33 94.02 21.43 0.61
iMM904 red linMOMA 437 9 31 33 93.38 21.43 0.6
iMM904 red MOMA 437 9 31 33 93.38 21.43 0.6
iLL672 full MOMA 433 9 35 33 92.52 21.43 0.57
Galactose iMM904 full FBA 595 32 36 59 94.29 35.16 0.58
iMM904 full linMOMA 595 32 36 59 94.29 35.16 0.58
iMM904 full MOMA 595 32 36 59 94.29 35.16 0.58
iMM904 red FBA 409 12 33 56 92.53 17.65 0.67
iMM904 red linMOMA 409 12 33 56 92.53 17.65 0.67
iMM904 red MOMA 409 12 33 56 92.53 17.65 0.67
iLL672 full MOMA 411 19 31 49 92.99 27.94 0.61
Glycerol iMM904 full FBA 596 43 36 47 94.3 47.78 0.48
iMM904 full linMOMA 595 44 37 46 94.15 48.89 0.47
iMM904 full MOMA 598 44 34 46 94.62 48.89 0.48
iMM904 red FBA 410 20 34 46 92.34 30.3 0.57
iMM904 red linMOMA 409 21 35 45 92.12 31.82 0.56
iMM904 red MOMA 412 21 32 45 92.79 31.82 0.57
iLL672 full MOMA 406 20 38 46 91.44 30.3 0.55
Ethanol iMM904 full FBA 593 45 29 55 95.34 45 0.54
iMM904 full linMOMA 592 45 30 55 95.18 45 0.54
iMM904 full MOMA 592 44 30 56 95.18 44 0.55
iMM904 red FBA 408 21 27 54 93.79 28 0.64
iMM904 red linMOMA 407 21 28 54 93.56 28 0.63
iMM904 red MOMA 407 20 28 55 93.56 26.67 0.64
iLL672 full MOMA 401 13 34 62 92.18 17.33 0.68
MCC, Matthews correlation coefficient (see Methods). Note that the iLL672 predictions were obtained directly from [3] and thus the viability threshold was not optimized using the maximum MCC approach.
Mo et al. BMC Systems Biology 2009 3:37  http://dx.doi.org:/10.1186/1752-0509-3-37

 

The values reported in Table 1 correspond to selecting

  • the optimal viability threshold based on this criterion.

We summarized the overall prediction accuracy of a model and method

  • using the Matthews Correlation Coefficient (MCC) [40].

The MCC ranges from -1 (all predictions incorrect) to +1 (all predictions correct) and

  • is suitable for summarizing overall prediction performance

in our case where there are substantially more viable than lethal gene deletions.

ROC plots were produced in Matlab (Mathworks, Inc.).

 

Table 1. Comparison of iMM904 and iLL672

  • gene deletion predictions and
  • experimental data

Inferring perturbed metabolic regions based on EM profiles

The method implemented in this study is shown schematically in Figures 1 and 2

Constraining the iMM904 network 

Relative levels of quantitative EM data were incorporated into the constraint-based framework

  • as overflow secretion exchange fluxes to simulate the required low-level production of
  • experimentally observed excreted metabolites.

The primary objective of this study is to associate

  • relative metabolite levels that are generally measured for metabonomic or biofluid analyses
  • to the quantitative ranges of intracellular reaction fluxes required to produce them.

However, without detailed kinetic information or dynamic metabolite measurements available,

  • we approximated EM datasets of relative quantitative metabolite levels
  • to be proportional to the rate in which they are secreted and detected
  • (at a steady state) – into the extracellular media.

This approach is analogous to approximating uptake rates based

  • on metabolite concentrations from a previous study performing sampling analysis
  • on a cardiomyocyte mitochondrial network
  • to identify differential flux distribution ranges

for various environmental (i.e. substrate uptake) conditions [19].

The raw data was normalized by the raw maximum value of the dataset
(thus the maximum secretion flux was 1 mmol/hr/gDW) with

  • an assumed error of 10%
  • to set the lower and upper bounds and thus
  • inherently accounting for sampling calculation sensitivity.

The gdh1/GDH2 strains were flask cultured under minimal glucose media conditions; thus,

  • glucose and oxygen uptake rates were set at 15 and 2 mmol/hr/gDW, respectively,
  • for the gdh1/GDH2 strain study.

In the anaerobic case the oxygen uptake rate was set to zero, and

  • sterols and fatty acids were provided as in silico supplements as described in [35].

For the potassium limitation/ammonium toxicity study

  • the growth rate was set at 0.17 1/h, and
  • the glucose uptake rate was minimized
  • to mimic experimental chemostat cultivation conditions.

These input constraints were constant for each perturbation and comparative wild-type condition

  • such that the calculated solution spaces between the conditions
  • differed based only on variations in the output secretion constraints.

FBA optimization of EM-constrained networks

A modified FBA method with minimization of the 1-norm objective function

  • between two optimal flux distributions was used
  • to determine optimal intracellular fluxes
  • based on the EM-constrained metabolic models.

This method determines two optimal flux distributions simultaneously

  • for two differently constrained models (e.g. wild type vs. mutant) –
  • these flux distributions maximize biomass production in each case and
  • the 1-norm distance between the distributions is as small as possible
  • given the two sets of constraints.

This approach avoids problems with

  • alternative optimal solutions when comparing two FBA-computed flux distributions
  • by assuming minimal rerouting of flux distibution between a perturbed network and its reference network.

Reaction flux changes from the FBA optimization results were determined

  • by computing the relative percentage fold change for each reaction
  • between the mutant and wild-type flux distributions.

Random sampling of the steady-state solution space

We utilized artificial centering hit-and-run (ACHR) Monte Carlo sampling [19,41]

  • to uniformly sample the metabolic flux solution space
  • defined by the constraints described above.

Reactions, and their participating metabolites, found to participate in intracellular loops [42]

  • were discarded from further analysis as these reactions can have arbitrary flux values.

The following sections describe the approaches used for the analysis of the different datasets.

Sampling approach used in the gdh1/GDH2 study

Due to the overall shape of the metabolic flux solution space,

  • most of the sampled flux distributions resided close to the minimally allowed growth rate
    (i.e. biomass production) and
  • corresponded to various futile cycles that utilized substrates but
  • did not produce significant biomass.

In order to study more physiologically relevant portions of the flux space

  • we restricted the sampling to the part of the solution space
  • where the growth rate was at least 50% of the maximum growth rate
  • for the condition as determined by FBA.

This assumes that cellular growth remains an important overall objective by the yeast cells

  • even in batch cultivation conditions, but
  • that the intracellular flux distributions
  • may not correspond to maximum biomass production [43].

To test the sensitivity of the results to the minimum growth rate threshold,

  • separate Monte Carlo samples were created for each minimum threshold
  • ranging from 50% to 100% at 5% increments.

We also tested the sensitivity of the results

  • to the relative magnitude of the extracellular metabolite secretion rates
  • by performing the sampling at three different relative levels

(0 corresponding to no extracellular metabolite secretion, maximum rate of 0.5 mmol/hr/gDW,
and maximum rate of 1.0 mmol/hr/gDW).

For each minimum growth rate threshold and extracellular metabolite secretion rate,

  • the ACHR sampler was run for 5 million steps and
  • a flux distribution was stored every 5000 steps.

The sensitivity analysis results are presented in Figures S3 and S4 [see Additional File 3], and

  • the results indicate that the reaction Z-scores (see below) are not significantly affected by
  1. either the portion of the solution space sampled or
  2. the exact scaling of secretion rates.

The final overall sample used was created by combining the samples for all minimum growth rate thresholds

  • for the highest extracellular metabolite secretion rate (maximum 1 mmol/hr/gDW).

This approach allowed biasing the sampling towards

  • physiologically relevant parts of the solution space
  • without imposing the requirement of strictly maximizing a predetermined objective function.

The samples obtained with no EM data were used as control samples

  • to filter reporter metabolites/subsystems whose scores were significantly high
  • due to only random differences between sampling runs.

Sampling approach used in the potassium limitation/ammonium toxicity study

Since the experimental data used in this study was generated in chemostat conditions, and

  • previous studies have indicated that chemostat flux patterns predicted by FBA are
  • close to the experimentally measured ones [43],
  • we assumed that sampling of the optimal solution space was appropriate for this study.

In order to sample a physiologically reasonable range of flux distributions,

  • samples for four different oxygen uptake rates
    (1, 2, 3, and 4 mmol/hr/gDW with 5 million steps each)
  • were combined in the final analysis.

Standardized scoring of flux differences between perturbation and control conditions

Z-score based approach was implemented to quantify differences in flux samples between two conditions (Figure 2).
First, two flux vectors were chosen randomly,

  • one from each of the two samples to be compared and
  • the difference between the flux vectors was computed.

This approach was repeated to create a sample of 10,000 (n) flux difference vectors

  • for each pair of conditions considered (e.g. mutant or perturbed environment vs. wild type).

Based on this flux difference sample, the sample mean (μdiff,i) and standard deviation (σdiff,i)

  • between the two conditions was calculated for each reaction i. The reaction Z-score was calculated as:

 

reaction Z-score

reaction Z-score

which describes the sampled mean difference deviation

  • from a population mean change of zero (i.e. no flux difference
    between perturbation and wild type).

Note that this approach allows accounting for uncertainty in the

  • flux distributions inferred based on the extracellular metabolite secretion constraints.

This is in contrast to approaches such as FBA or MoMA that would predict

  • a single flux distribution for each condition and thus potentially
  • overestimate differences between conditions.

The reaction Z-scores can then be further used in analysis

  • to identify significantly perturbed regions of the metabolic network
  • based on reporter metabolite [44] or subsystem [30] Z-scores.

These reporter regions indicate, or “report”, dominant perturbation features

  • at the metabolite and pathway levels for a particular condition.

The reporter metabolite Z-score for any metabolite can be derived from the reaction Z-scores

  • of the reactions consuming or producing j (set of reactions denoted as Rj) as:

 

reporter z-score for any metabolite j

reporter z-score for any metabolite j

where Nis the number of reactions in Rand mmet,is calculated as

 

distributional correction for m_met,j SQRT

distributional correction for m_met,j SQRT

To account and correct for background distribution, the metabolite Z-score was normalized

  • by computing μmet,Nj and σmet,,Nj corresponding to the mean mmet and
  • its standard deviation for 1,000 randomly generated reaction sets of size Nj.

Z-scores for subsystems were calculated similarly by considering the set of reactions R

  • that belongs to each subsystem k.

Hence, positive metabolite and subsystem scores indicate a significantly perturbed metabolic region

  • relative to other regions, whereas
  • a negative score indicate regions that are not perturbed
  • more significantly than what is expected by random chance.

Perturbation subnetworks of reactions and connecting metabolites were visualized using Cytoscape [45].

Results and discussion

  1. Reconstruction and validation of iMM904 network iMM904 network content 

A previously reconstructed S. cerevisiae network, iND750,

  • was used as the basis for the construction of the expanded iMM904 network.
  • Prior to its presentation here, the
    iMM904 network content was the basis for a consensus jamboree network that was recently published
  • but has not yet been adapted for FBA calculations [46].

The majority of iND750 content was carried over and

  • further expanded on to construct iMM904, which accounts for
  1. 904 genes,
  2. 1,228 individual metabolites, and
  3. 1,412 reactions of which
  •                       395 are transport reactions.

Both the number of gene-associated reactions and the number of metabolites

  • increased in iMM904 compared with the iND750 network.

Additional genes and reactions included in the network primarily expanded the

  • lipid,
  • transport, and
  • carbohydrate subsystems.

The lipid subsystem includes

  • new genes and
  • reactions involving the degradation of sphingolipids and glycerolipids.

Sterol metabolism was also expanded to include

  • the formation and degradation of steryl esters, the
  •                      storage form of sterols.

The majority of the new transport reactions were added

  • to connect network gaps between intracellular compartments
  • to enable the completion of known physiological functions.

We also added a number of new secretion pathways

  • based on experimentally observed secreted metabolites [31].

A number of gene-protein-reaction (GPR) relationships were modified

  • to include additional gene products that are required to catalyze a reaction.

For example, the protein compounds

  • thioredoxin and
  • ferricytochrome C

were explicitly represented as compounds in iND750 reactions, but

  • the genes encoding these proteins were not associated with their corresponding GPRs.

Other examples include glycogenin and NADPH cytochrome p450 reductases (CPRs),

  1. which are required in the assembly of glycogen and
  2. to sustain catalytic activity in cytochromes p450, respectively.

These additional proteins were included in iMM904 as

  • part of protein complexes to provide a more complete
  • representation of the genes and
  • their corresponding products necessary for a catalytic activity to occur.

Major modifications to existing reactions were in cofactor biosynthesis, namely in

  • quinone,
  • beta-alanine, and
  • riboflavin biosynthetic pathways.

Reactions from previous S. cerevisiae networks associated with

  • quinone,
  • beta-alanine, and
  • riboflavin biosynthetic pathways

were essentially inferred from known reaction mechanisms based on

  • reactions in previous network reconstructions of E. coli [2,47].

These pathways were manually reviewed

  • based on current literature and subsequently replaced by
  • reactions and metabolites specific to yeast.

Additional changes in other subsystems were also made, such as

  1. changes to the compartmental location of a gene and
  2. its corresponding reaction(s),
  3. changes in reaction reversibility and cofactor specificity, and
  4. the elucidation of particular transport mechanisms.

A comprehensive listing of iMM904 network contents as well as

  • a detailed list of changes between iND750 and iMM904 is included
    [see Additional file 1].

Predicting deletion growth phenotypes

The updated genome-scale iMM904 metabolic network was validated

  • by comparing in silico single-gene deletion predictions to
  • in vivo results from a previous study used
  • to analyze another S. cerevisiae metabolic model, iLL672 [3].

This network was constructed based on the iFF708 network [22],

  • which was also the starting point for
  • reconstructing the iND750 network [2].

The experimental data used to validate the iLL672 model consisted of

3,360 single-gene knockout strain phenotypes evaluated

  • under minimal media growth conditions with
  1. glucose,
  2. galactose,
  3. glycerol, and
  4. ethanol

as sole carbon sources. Growth phenotypes for the iMM904 network were predictedusing

  1. FBA [3234],
  2. MoMA [35], and
  3. linear MoMA methods

as described in Methods and subsequently compared to the experimental data (Table 1).

Each deleted gene growth prediction comparison was classified as

  1. true lethal,
  2. true viable,
  3. false lethal, or
  4. false viable.

The growth rate threshold for considering a prediction viable was chosen

  • for each condition and method separately
  • to optimize the tradeoff between true viable and false viable predictions
    (maximum Matthews correlation coefficient, see Methods).

Since iMM904 has 212 more genes than iLL672 with experimental data, we also present results

  • for the subset of iMM904 predictions with genes included in iLL672 (reduced iMM904 set).

When the same gene sets are compared, iMM904 improves gene lethality predictions under

  • glucose,
  • galactose, and
  • glycerol conditions

over iLL672 somewhat, but is less accurate

  • at predicting growth phenotypes under the ethanol condition.

It should be noted that the iLL672 predictions were obtained directly from [3]

  • thus the growth rate threshold was not optimized similarly to iMM904 predictions.

Overall, when viability cutoff is chosen

  • as indicated above for each method separately,
  • the three prediction methods perform similarly
  1. FBA,
  2. MOMA, and
  3. linear MOMA) .

While the full gene complement in iMM904 greatly increased

  • the number of true viable predictions,
  • the full model also made significantly more false viable predictions
  • compared with reduced iMM904 and iLL672 predictions.

However, it is important to note that 143 reactions involved in dead-end biosynthetic pathways were actually

  • removed from iFF708 to build the iLL672 reconstruction [3].

These dead-ends are considered “knowledge gaps” in pathways

  • that have not been fully characterized and, as a result,
  • lead to false viable predictions when determining gene essentiality
  • if the pathway is in fact required for growth under a certain condition [2,26].

As more of these pathways are elucidated and

  • included in the model to
  • fill in existing network gaps,
  • we can expect false viable prediction rates to consequently decrease.

Thus, while a larger network has a temporarily reduced capacity to accurately predict gene deletion phenotypes,

  • it captures a more complete picture of currently known metabolic functions and
  • provides a framework for network expansion as new pathways are elucidated [48].

 

Inferring intracellular perturbation states from metabolic profiles – Aerobic and anaerobic gdh1/GDH2 mutant behavior

The gdh1/GDH2 mutant strain was previously developed [49,50]

  • to lower NADPH consumption in ammonia assimilation, which would
  • favor the NADPH-dependent fermentation of xylose.

In this strain, the NADPH-dependent glutamate dehydrogenase, Gdh1, was

  • deleted and the NADH-dependent form of the enzyme, Gdh2,
  •                     was overexpressed.

The net effect is to allow efficient assimilation of ammonia

  • into glutamate using NADH instead of NADPH as a cofactor.

While growth characteristics remained unaffected,

  • relative quantities of secreted metabolites differed between the wild-type and mutant strain
  • under aerobic and anaerobic conditions.

We analyzed EM data for the gdh1/GDH2 and wild-type strains reported

  • in [31] under aerobic and anaerobic conditions separately using
  • both FBA optimization and
  • sampling-based approaches as described in Methods.

43 measured extracellular and intracellular metabolites from the original dataset [31],

  • primarily of central carbon and amino acid metabolism,
  • were explicitly represented in the iMM904 network [see Additional file 4].

Extracellular metabolite levels were used

  • to formulate secretion constraints and
  • differential intracellular metabolites were used
  • to compare and validate the intracellular flux predictions.

Perturbed reactions from the FBA results were

  • determined by calculating relative flux changes, and
  • reaction Z-scores were calculated from the sampling analysis
  • to quantify flux changes between the mutant and wild-type strains,
  • with Z reaction > 1.96 corresponding to a two-tailed p-value < 0.05 and
  • considered to be significantly perturbed [see Additional file 4].

Additional file 4. Gdh mutant aerobic and anaerobic analysis results. 

The data provided are the full results for the exometabolomic analysis of aerobic and anerobic gdh1/GDH2 mutant.

Format: XLS Size: 669KB Download file

This file can be viewed with: Microsoft Excel Viewer

To validate the predicted results, reaction flux changes from both FBA and sampling methods were compared to differential intracellular metabolite level data measured from the same study. Intracellular metabolites involved in highly perturbed reactions (i.e. reactants and products) predicted from FBA and sampling analyses were identified and
compared to metabolites that were experimentally identified as significantly changed (< 0.05) between mutant and wild-type. Statistical measures of recall, accuracy, and
precision were calculated and represent the predictive sensitivity, exactness, and reproducibility respectively. From the sampling analysis, a considerably larger number of
significantly perturbed reactions are predicted in the anaerobic case (505 reactions, or 70.7% of active reactions) than in aerobic (394 reactions, or 49.8% of active reactions). The top percentile of FBA flux changes equivalent to the percentage of significantly perturbed sampling reactions were compared to the intracellular data. Results from both analyses are summarized in Table 2. Sampling predictions were considerably higher in recall than FBA predictions for both conditions, with respective ranges of 0.83–1
compared to 0.48–0.96. Accuracy was also higher in sampling predictions; however, precision was slightly better in the FBA predictions as expected due to the smaller
number of predicted changes. Overall, the sampling predictions of perturbed intracellular metabolites are strongly consistent with the experimental data and significantly
outperforms that of FBA optimization predictions in accurately predicting differential metabolites involved in perturbed intracellular fluxes.

Table 2. Statistical comparison of the differential intracellular metabolite data set (< 0.05) with metabolites involved in perturbed reactions predicted by FBA optimization and sampling analyses for aerobic and anaerobic gdh1/GDH2 mutant.

 

Table 2 Statistical comparison of the differential intracellular metabolite data set (p < 0.05)
with metabolites involved in perturbed reactions predicted by FBA optimization and
sampling analyses for aerobic and anaerobic gdh1/GDH2 mutant.
                           Aerobic                         Anaerobic                             Overall
FBA Sampling FBA Sampling FBA
Recall 0.48 0.83 0.96 1 0.71 0.91
Accuracy 0.55 0.62 0.64 0.64 0.6 0.63
Precision 0.78 0.69 0.64 0.63 0.68 0.66
Overall statistics indicate combined results of both conditions.
Mo et al. BMC Systems Biology 2009 3:37   http://dx.doi.org:/10.1186/1752-0509-3-37


Figure 3.
 Perturbation reaction subnetwork of gdh1/GDH2 mutant under aerobic conditions.

The network illustrates a simplified subset of highly perturbedPerturbation subnetworks can be drawn to visualize predicted significantly perturbed intracellular reactions and illustrate their connection to the observed secreted metabolites in the aerobic and anaerobic gdh1/GDH2 mutants.

Perturbation reaction subnetwork of gdh1.GDH2 mutant under aerobic conditions.

Perturbation reaction subnetwork of gdh1.GDH2 mutant under aerobic conditions.

Figure 3 shows an example of a simplified aerobic perturbation subnetwork consisting primarily of proximal pathways connected directly to a subset of major secreted
metabolites

  • glutamate,
  • proline,
  • D-lactate, and
  • 2-hydroxybuturate.

Figure 4 displays anaerobic reactions with Z-scores of similar magnitude to the perturbed reactions in Figure 3. The same subset of metabolites is also present in the
larger anaerobic perturbation network and indicates that the NADPH/NADH balance perturbation induced by the gdh1/GDH2 manipulation has widespread effects
beyond just altering glutamate metabolism anaerobically.

Interestingly, it is clear that the majority of the secreted metabolite pathways involve connected perturbed reactions that broadly converge on glutamate.

Note that Figures 3 and 4 only show the subnetworks that consisted of two or more connected reactions  for a number of secreted metabolites no contiguous perturbed pathway could be identified by the sampling approach. This indicates that the secreted metabolite pattern alone is not sufficient to determine which specific
production and secretion pathways are used by the cell for these metabolites.

Reactions connected to aerobically-secreted metabolites predicted from the sampling analysis of the gdh1/GDH2 mutant strain.
The major secreted metabolites

  • glutamate,
  • proline,
  • D-lactate, and
  • 2-hydroxybuturate

were also detected in the anaerobic condition. Metabolite abbreviations are found in Additional file 1.

Figure 4.

Perturbation reaction subnetwork of gdh1/GDH2 mutant under anaerobic conditions.

Perturbation reaction subnetwork of gdh1.GDH2 mutant under anaerobic conditions

Perturbation reaction subnetwork of gdh1.GDH2 mutant under anaerobic conditions

Subnetwork illustrates the highly perturbed anaerobic reactions of similar Z-reaction magnitude to the reactions in Figure 3.

A significantly larger number of reactions indicates mutant metabolic effects are more widespread in the anaerobic environment.
The network shows that perturbed pathways converge on glutamate, the main site in which the gdh1/GDH2 modification was introduced, which
suggests that the direct genetic perturbation effects are amplified under this environment. Metabolite abbreviations are found in Additional file 1.

To further highlight metabolic regions that have been systemically affected by the gdh1/GDH2 modification, reporter metabolite and subsystem methods [30] were used to
summarize reaction scores around specific metabolites and in specific metabolic subsystems. The top ten significant scores for metabolites/subsystems associated with more
than three reactions are summarized in Tables 3 (aerobic) and 4 (anaerobic), with Z > 1.64 corresponding to < 0.05 for a one-tailed distribution. Full data for all reactions,
reporter metabolites, and reporter subsystems is included [see Additional file 4].

Table 3. List of the top ten significant reporter metabolite and subsystem scores for the gdh1/GDH2 vs. wild type comparison in aerobic conditions.

Table 3
List of the top ten significant reporter metabolite and subsystem scores for the gdh1/GDH2 vs. wild type comparison in aerobic conditions.
Reporter metabolite Z-score No of reactions*
L-proline [c] 2.71 4
Carbon dioxide [m] 2.51 15
Proton [m] 2.19 51
Glyceraldehyde 3-phosphate [c] 1.93 7
Ubiquinone-6 [m] 1.82 5
Ubiquinol-6 [m] 1.82 5
Ribulose-5-phosphate [c] 1.8 4
Uracil [c] 1.74 4
L-homoserine [c] 1.72 4
Alpha-ketoglutarate [m] 1.71 8
Reporter subsystem Z-score No of reactions
Citric Acid Cycle 4.58 7
Pentose Phosphate Pathway 3.29 12
Glycine and Serine Metabolism 2.69 17
Alanine and Aspartate Metabolism 2.65 6
Oxidative Phosphorylation 1.79 8
Thiamine Metabolism 1.54 8
Arginine and Proline Metabolism 1.44 20
Other Amino Acid Metabolism 1.28 5
Glycolysis/Gluconeogenesis 0.58 14
Anaplerotic reactions 0.19 9
*Number of reactions categorized in a subsystem or found to be neighboring each metabolite
Mo et al. BMC Systems Biology 2009 3:37   http://dx.doi.org:/10.1186/1752-0509-3-37

Table 4. List of top ten significant reporter metabolite and subsystem scores for the gdh1/GDH2 vs. wild type comparison in anaerobic conditions.

 

Table 4
List of top ten significant reporter metabolite and subsystem scores for the gdh1/GDH2 vs. wild type comparison in anaerobic conditions.
Reporter metabolite Z-score No of reactions
Glutamate [c] 4.52 35
Aspartate [c] 3.21 11
Alpha-ketoglutarate [c] 2.66 17
Glycine [c] 2.65 7
Pyruvate [m] 2.56 7
Ribulose-5-phosphate [c] 2.43 4
Threonine [c] 2.28 6
10-formyltetrahydrofolate [c] 2.27 5
Fumarate [c] 2.27 5
L-proline [c] 2.04 4
Reporter subsystem Z-score No of reactions
Valine, Leucine, and Isoleucine Metabolism 3.97 15
Tyrosine, Tryptophan, and Phenylalanine Metabolism 3.39 23
Pentose Phosphate Pathway 3.29 11
Purine and Pyrimidine Biosynthesis 3.08 40
Arginine and Proline Metabolism 2.96 19
Threonine and Lysine Metabolism 2.74 14
NAD Biosynthesis 2.66 7
Alanine and Aspartate Metabolism 2.65 6
Histidine Metabolism 2.24 10
Cysteine Metabolism 1.85 10
Mo et al. BMC Systems Biology 2009 3:37   http://dx.doi.org:/10.1186/1752-0509-3-37
Open Data

Perturbations under aerobic conditions largely consisted of pathways involved in mediating the NADH and NADPH balance. Among the highest scoring aerobic subsystems
are TCA cycle and pentose phosphate pathway – key pathways directly involved in the generation of NADH and NADPH. Reporter metabolites involved in these
subsystems –

  • glyceraldehyde-3-phosphate,
  • ribulose-5-phosphate, and
  • alpha-ketoglutarate – were also identified.

These results are consistent with flux and enzyme activity measurements

  • of the gdh1/GDH2 strain under aerobic conditions [32],
  1. which reported significant reduction in the pentose phosphate pathway flux
  2. with concomitant changes in other central metabolic pathways.

Levels of several TCA cycle intermediates (e.g. fumarate, succinate, malate) were also elevated

  • in the gdh1/GDH2 mutant according to the differential intracellular metabolite data.

Altered energy metabolism, as indicated by

  • reporter metabolites (i.e. ubiquinone- , ubiquinol, mitochondrial proton)
  • and subsystem (oxidative phosphorylation),

is certainly feasible as NADH is a primary reducing agent for ATP production.

Pentose phosphate pathway and NAD biosynthesis also appears

  • among the most perturbed anaerobic subsystems, further suggesting
  • perturbed cofactor balance as a common, dominant effect under both conditions.

Glutamate dehydrogenase is a critical enzyme of amino acid biosynthesis as it acts as

  • the entry point for ammonium assimilation via glutamate.

Consequently, metabolic subsystems involved in amino acid biosynthesis were broadly perturbed

  • as a result of the gdh1/GDH2 modification in both aerobic and anaerobic conditions.

For example, the proline biosynthesis pathway that uses glutamate as a precursor

  • was significantly perturbed in both conditions,
  • with significantly changed intracellular and extracellular levels.

There were differences, however, in that more amino acid related subsystems were

  • significantly affected in the anaerobic case (Table 4),
  • further highlighting that altered ammonium assimilation in the mutant
  • has a more widespread effect under anaerobic conditions.

This effect is especially pronounced for

  • threonine and nucleotide metabolism,
  • which were predicted to be significantly perturbed only in anaerobic conditions.

Intracellular threonine levels were amongst the most significantly reduced

  • relative to other differential intracellular metabolites in the anaerobically grown gdh1/GDH2 strain
    (see [31] and Additional file 4), and
  • the relationship between threonine and nucleotide biosynthesis is further supported

by threonine’s recently discovered role as a key precursor in yeast nucleotide biosynthesis [51].

Other key anaerobic reporter metabolites are

  • glycine and 10-formyltetrahydrofolate,
  • both of which are involved in the cytosolic folate cycle (one-carbon metabolism).

Folate is intimately linked to biosynthetic pathways of

  • glycine (with threonine as its precursor) and purines
  • by mediating one-carbon reaction transfers necessary in their metabolism and
  • is a key cofactor in cellular growth [52].

Thus, the anaerobic perturbations identified in the analysis emphasize the close relationship

  • between threonine, folate, and nucleotide metabolic pathways as well as
  • their potential connection to perturbed ammonium assimilation processes.

Interestingly, this association has been previously demonstrated at the transcriptional level

  • as yeast ammonium assimilation (via glutamine synthesis) was found to be
  • co-regulated with genes involved in glycine, folate, and purine synthesis [53].

In summary, the overall differences in predicted gdh1/GDH2 mutant behavior

  • under aerobic and anaerobic conditions show that changes in flux states
  • directly related to modified ammonium assimilation pathway
  1. are amplified anaerobically whereas the
  2. indirect effects through NADH/NADPH balance are more significant aerobically.

Perturbed metabolic regions under aerobic conditions were predominantly

  • in central metabolic pathways involved in responding to the changed NADH/NADPH demand
  • and did not necessarily emphasize that glutamate dehydrogenase was the site of the genetic modification.

The majority of affected anaerobic pathways were involved directly

  • in modified ammonium assimilation as evidenced by

1) significantly perturbed amino acid subsystems,

2) a broad perturbation subnetwork converging on glutamate (Figure 4), and

3) glutamate as the most significant reporter metabolite (Table 4).

Potassium-limited and excess ammonium environments

A recent study reported that potassium limitation resulted in significant

  • growth retardation effect in yeast due to excess ammonium uptake
  • when ammonium was provided as the sole nitrogen source [33].

The proposed mechanism for this effect was that ammonium

  • could to be freely transported through potassium channels
  • when potassium concentrations were low in the media environment, thereby
  • resulting in excess ammonium uptake [33].

As a result, yeast incurred a significant metabolic cost

  • in assimilating ammonia to glutamate and
  • secreting significant amounts of glutamate and other amino acids
  • in potassium-limited conditions as a means to detoxify the excess ammonium.

A similar effect was observed when yeast was grown

  • with no potassium limitation,
  • but with excess ammonia in the environment.

While the observed effect of both environments (low potassium or excess ammonia) was similar,

  • quantitatively unique amino acid secretion profiles suggested that
  • internal metabolic states in these conditions are potentially different.

In order to elucidate the differences in internal metabolic states, we utilized

  • the iMM904 model and the EM profile analysis method to analyze amino acid secretion profiles
  • for a range of low potassium and high ammonia conditions reported in [33].

As before, we utilized amino acid secretion patterns as constraints to the iMM904 model,

  1. sampled the allowable solution space,
  2. computed reaction Z-scores for changes from a reference condition (normal potassium and ammonia), and
  3. finally summarized the resulting changes using reporter metabolites.

Figure 5 shows a clustering of the most significant reporter metabolites (Z ≥ 1.96 in any of the four conditions studied)

  • obtained from this analysis across the four conditions studied.

Interestingly, the potassium-limited environment perturbed only a subset of

  • the significant reporter metabolites identified in the high ammonia environments.

Both low potassium environments shared a consistent pattern of

  • highly perturbed amino acids and related precursor biosynthesis metabolites
    (e.g. pyruvate, PRPP, alpha-ketoglutarate)
  • with high ammonium environments.

The amino acid perturbation pattern (indicated by red labels in Figure 5) was present in

  • the ammonium-toxic environments, although the pattern was
  • slightly weaker for the lower ammonium concentration.

Nevertheless, the results clearly indicate that a similar

  • ammonium detoxifying mechanism that primarily perturbs pathways
  • directly related to amino acid metabolism
  • exists under both types of media conditions.

Figure 5.

Clustergram of top reporter metabolites - y in ammonium-toxic and potassium-limited conditions

Clustergram of top reporter metabolites – y in ammonium-toxic and potassium-limited conditions

Clustergram of top reporter metabolites (i.e. in yellow) in ammonium-toxic and potassium-limited conditions.

Amino acid perturbation patterns (shown in red) were shown to be consistently scored across conditions, indicating that potassium-limited environments K1 (lowest
concentration) and K2 (low concentration) elicited a similar ammonium detoxification response as ammonium-toxic environments N1 (high concentration) and N2
(highest concentration). Metabolites associated with folate metabolism (highlighted in green) are also highly perturbed in ammonium-toxic conditions. Metabolite
abbreviations are found in Additional file 1.

In addition to perturbed amino acids, a secondary effect notably appears at high ammonia levels in which metabolic regions related to folate metabolism are significantly affected. As highlighted in green in Figure 3, we predicted significantly perturbed key metabolites involved in the cytosolic folate cycle. These include tetrahydrofolate derivatives and other metabolites connected to the folate pathway, namely glycine and the methionine-derived methylation cofactors S-adenosylmethionine and S-adenosyl-homocysteine. Additionally, threonine was identified to be a key perturbed metabolite in excess ammonium conditions. These results further illustrate the close
connection between threonine biosynthesis, folate metabolism involving glycine derived from its threonine precursor, and nucleotide biosynthesis [51] that was discussed in
conjunction with the gdh1/GDH2 strain data. Taken together with the anaerobic gdh1/GDH2 data, the results consistently suggest highly perturbed threonine and folate
metabolism when amino acid-related pathways are broadly affected.

In both ammonium-toxic and potassium-limited environments, impaired cellular growth was observed, which can be attributed to high energetic costs of increased
ammonium assimilation to synthesize and excrete amino acids. However, under high ammonium environments, reporter metabolites related to threonine and folate
metabolism indicated that their perturbation, and thus purine supply, may be an additional factor in decreasing cellular viability as there is a direct relationship between
intracellular folate levels and growth rate [54]. Based on these results, we concluded that while potassiumlimited growth in yeast indeed shares physiological features with
growth in ammonium excess, its effects are not as detrimental as actual ammonium excess. The effects on proximal amino acid metabolic pathways are similar in both
environments as indicated by the secretion of the majority of amino acids. However, when our method was applied to analyze the physiological basis behind differences in
secretion profiles between low potassium and high ammonium conditions, ammonium excess was predicted to likely disrupt physiological ammonium assimilation processes,
which in turn potentially impacts folate metabolism and associated cellular growth.

Conclusion

The method presented in this study presents an approach to connecting intracellular flux states to metabolites that are excreted under various physiological conditions. We
showed that well-curated genome-scale metabolic networks can be used to integrate and analyze quantitative EM data by systematically identifying altered intracellular
pathways related to measured changes in the extracellular metabolome. We were able to identify statistically significant metabolic regions that were altered as a result of
genetic (gdh1/GD2 mutant) and environmental (excess ammonium and limited potassium) perturbations, and the predicted intracellular metabolic changes were consistent
with previously published experimental data including measurements of intracellular metabolite levels and metabolic fluxes. Our reanalysis of previously published EM data
on ammonium assimilation-related genetic and environmental perturbations also resulted in testable hypotheses about the role of threonine and folate pathways in mediating
broad responses to changes in ammonium utilization. These studies also demonstrated that the samplingbased method can be readily applied when only partial secreted
metabolite profiles (e.g. only amino acids) are available.

With the emergence of metabolite biofluid biomarkers as a diagnostic tool in human disease [55,56] and the availability of genome-scale human metabolic networks [1],
extensions of the present method would allow identifying potential pathway changes linked to these biomarkers. Employing such a method for studying yeast metabolism was possible as the metabolomic data was measured under controllable environmental conditions where the inputs and outputs of the system were defined. Measured metabolite biomarkers in a clinical setting, however, is far from a controlled environment with significant variations in genetic, nutritional, and environmental factors between different
patients. While there are certainly limitations for clinical applications, the method introduced here is a progressive step towards applying genome-scale metabolic networks
towards analyzing biofluid metabolome data as it 1) avoids the need to only study optimal metabolic states based on a predetermined objective function, 2) allows dealing with noisy experimental data through the sampling approach, and 3) enables analysis even with limited identification of metabolites in the data. The ability to establish potential
connections between extracellular markers and intracellular pathways would be valuable in delineating the genetic and environmental factors associated with a particular
disease.

Authors’ contributions

Conceived and designed the experiments: MLM MJH BOP. Performed experiments: MLM MJH. Analyzed the data: MLM MJH. Wrote the paper: MLM MJH BOP. All authors have read and approved the final manuscript.

Acknowledgements

We thank Jens Nielsen for providing the raw metabolome data for the mutant strain, and Jan Schellenberger and Ines Thiele for valuable discussions. This work was supported by NIH grant R01 GM071808. BOP serves on the scientific advisory board of Genomatica Inc.

 

References

  1. Duarte NC, Becker SA, Jamshidi N, Thiele I, Mo ML, Vo TD, Srivas R, Palsson BO: Global reconstruction of the human metabolic network based on genomic and bibliomic data. 

Proc Natl Acad Sci USA 2007, 104(6):1777-1782. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Duarte NC, Herrgard MJ, Palsson B: Reconstruction and Validation of Saccharomyces cerevisiae iND750, a Fully Compartmentalized Genome-Scale Metabolic Model. 

Genome Res 2004, 14(7):1298-1309. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Kuepfer L, Sauer U, Blank LM: Metabolic functions of duplicate genes in Saccharomyces cerevisiae. 

Genome Res 2005, 15(10):1421-1430. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Nookaew I, Jewett MC, Meechai A, Thammarongtham C, Laoteng K, Cheevadhanarak S, Nielsen J, Bhumiratana S: The genome-scale metabolic model iIN800 of Saccharomyces cerevisiae and its validation: a scaffold to query lipid metabolism. 

BMC Syst Biol 2008, 2:71. PubMed Abstract | BioMed Central Full Text | PubMed Central Full Text

  1. Edwards JS, Palsson BO: Systems properties of the Haemophilus influenzae Rd metabolic genotype. 

J biol chem 1999, 274(25):17410-17416. PubMed Abstract | Publisher Full Text

  1. Edwards JS, Palsson BO: The Escherichia coli MG1655 in silico metabolic genotype: Its definition, characteristics, and capabilities. 

Proc Natl Acad Sci USA 2000, 97(10):5528-5533. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Thiele I, Vo TD, Price ND, Palsson B: An Expanded Metabolic Reconstruction of Helicobacter pylori (IT341 GSM/GPR): An in silico genome-scale characterization of single and double deletion mutants. 

J Bacteriol 2005, 187(16):5818-5830. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Vo TD, Greenberg HJ, Palsson BO: Reconstruction and functional characterization of the human mitochondrial metabolic network based on proteomic and biochemical data. 

J Biol Chem 2004, 279(38):39532-39540. PubMed Abstract | Publisher Full Text

  1. Reed JL, Vo TD, Schilling CH, Palsson BO: An expanded genome-scale model of Escherichia coli K-12 (iJR904 GSM/GPR). 

Genome Biology 2003, 4(9):R54.51-R54.12. BioMed Central Full Text

  1. Feist AM, Henry CS, Reed JL, Krummenacker M, Joyce AR, Karp PD, V H, Palsson BO: A genome-scale metabolic reconstruction for Escherichia coli K-12 MG1655 that accounts for 1261 ORFs and thermodynamic information. 

Molecular Systems Biology 2007, 3:121. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Suthers PF, Dasika MS, Kumar VS, Denisov G, Glass JI, Maranas CD: A genome-scale metabolic reconstruction of Mycoplasma genitalium, iPS189. 

PLos Comp Biol 2009, 5(2):e1000285. Publisher Full Text

  1. Price ND, Reed JL, Palsson BO: Genome-scale models of microbial cells: evaluating the consequences of constraints. 

Nat Rev Microbiol 2004, 2(11):886-897. PubMed Abstract | Publisher Full Text

  1. Becker SA, Feist AM, Mo ML, Hannum G, Palsson BO, Herrgard MJ: Quantitative Prediction of Cellular Metabolism with Constraint-based Models: The COBRA Toolbox. 

Nature protocols 2007, 2(3):727-738. PubMed Abstract | Publisher Full Text

  1. Reed JL, Palsson BO: Genome-Scale In Silico Models of E. coli Have Multiple Equivalent Phenotypic States: Assessment of Correlated Reaction Subsets That Comprise Network States. 

Genome Res 2004, 14(9):1797-1805. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Fong SS, Palsson BO: Metabolic gene deletion strains of Escherichia coli evolve to computationally predicted growth phenotypes. 

Nature Genetics 2004, 36(10):1056-1058. PubMed Abstract | Publisher Full Text

  1. Ibarra RU, Edwards JS, Palsson BO: Escherichia coli K-12 undergoes adaptive evolution to achieve in silico predicted optimal growth. 

Nature 2002, 420(6912):186-189. PubMed Abstract | Publisher Full Text

  1. Schellenberger J, Palsson BØ: Use of randomized sampling for analysis of metabolic networks. 

J Biol Chem 2009, 284(9):5457-5461. PubMed Abstract | Publisher Full Text

  1. Almaas E, Kovács B, Vicsek T, Oltvai ZN, Barabási AL: Global organization of metabolic fluxes in the bacterium Escherichia coli. 

Nature 2004, 427(6977):839-843. PubMed Abstract | Publisher Full Text

  1. Thiele I, Price ND, Vo TD, Palsson BO: Candidate metabolic network states in human mitochondria: Impact of diabetes, ischemia, and diet. 

J Biol Chem 2005, 280(12):11683-11695. PubMed Abstract | Publisher Full Text

  1. Kell DB: Metabolomics and systems biology: making sense of the soup. 

Curr Opin Microbiol 2004, 7(3):296-307. PubMed Abstract | Publisher Full Text

  1. Kell DB, Brown M, Davey HM, Dunn WB, Spasic I, Oliver SG: Metabolic footprinting and systems biology: the medium is the message. 

Nat Rev Microbiol 2005, 3(7):557-565. PubMed Abstract | Publisher Full Text

  1. Goodacre R, Vaidyanathan S, Dunn WB, Harrigan GG, Kell DB: Metabolomics by numbers: acquiring and understanding global metabolite data. 

Trends Biotechnol 2004, 22(5):245-252. PubMed Abstract | Publisher Full Text

  1. Lenz EM, Bright J, Wilson ID, Morgan SR, Nash AF: A 1H NMR-based metabonomic study of urine and plasma samples obtained from healthy human subjects. 

J Pharm Biomed Anal 2003, 33(5):1103-1115. PubMed Abstract | Publisher Full Text

  1. Allen J, Davey HM, Broadhurst D, Heald JK, Rowland JJ, Oliver SG, Kell DB: High-throughput classification of yeast mutants for functional genomics using metabolic footprinting. 

Nat Biotech 2003, 21(6):692-696. Publisher Full Text

  1. Nicholson JK, Connelly J, Lindon JC, Holmes E: Metabonomics: a platform for studying drug toxicity and gene function. 

Nat Rev Drug Discov 2002, 1(2):153-161. PubMed Abstract | Publisher Full Text

  1. Mortishire-Smith RJ, Skiles GL, Lawrence JW, Spence S, Nicholls AW, Johnson BA, Nicholson JK: Use of metabonomics to identify impaired fatty acid metabolism as the mechanism of a drug-induced toxicity. 

Chem Res Toxicol 2004, 17(2):165-173. PubMed Abstract | Publisher Full Text

  1. Sabatine MS, Liu E, Morrow DA, Heller E, McCarroll R, Wiegand R, Berriz GF, Roth FP, Gerszten RE: Metabolomic identification of novel biomarkers of myocardial ischemia. 

Circulation 2005, 112(25):3868-3875. PubMed Abstract | Publisher Full Text

  1. Cakir T, Efe C, Dikicioglu D, Hortaçsu AKB, Oliver SG: Flux balance analysis of a genome-scale yeast model constrained by exometabolomic data allows metabolic system identification of genetically different strains. 

Biotechnol Prog 2007, 23(2):320-326. PubMed Abstract | Publisher Full Text

  1. Bang JW, Crockford DJ, Holmes E, Pazos F, Sternberg MJ, Muggleton SH, Nicholson JK: Integrative top-down system metabolic modeling in experimental disease states via data-driven Bayesian methods. 

J Proteome Res 2008, 7(2):497-503. PubMed Abstract | Publisher Full Text

  1. Oliveira AP, Patil KR, Nielsen J: Architecture of transcriptional regulatory circuits is knitted over the topology of bio-molecular interaction networks. 

BMC Syst Biol 2008, 2:17. PubMed Abstract | BioMed Central Full Text | PubMed Central Full Text

  1. Villas-Boas SG, Moxley JF, Akesson M, Stephanopoulos G, Nielsen J: High-throughput metabolic state analysis: the missing link in integrated functional genomics of yeasts. 

Biochem J 2005, 388(Pt 2):669-677. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Moreira dos Santos M, Thygesen G, Kötter P, Olsson L, Nielsen J: Aerobic physiology of redox-engineered Saccharomyces cerevisiae strains modified in the ammonium assimilation for increased NADPH availability. 

FEMS Yeast Res 2003, 4(1):59-68. PubMed Abstract | Publisher Full Text

  1. Hess DC, Lu W, Rabinowitz JD, Botstein D: Ammonium toxicity and potassium limitation in yeast. 

PLoS Biol 2006, 4(11):e351. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Nissen TL, Schulze U, Nielsen J, Villadsen J: Flux distributions in anaerobic, glucose-limited continuous cultures of Saccharomyces cerevisiae. 

Microbiology 1997, 143(Pt 1):203-218. PubMed Abstract | Publisher Full Text

  1. Famili I, Forster J, Nielsen J, Palsson BO: Saccharomyces cerevisiae phenotypes can be predicted by using constraint-based analysis of a genome-scale reconstructed metabolic network. 

Proc Natl Acad Sci USA 2003, 100(23):13134-13139. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Bonarius HPJ, Schmid G, Tramper J: Flux analysis of underdetermined metabolic networks: The quest for the missing constraints. 

Trends in Biotechnology 1997, 15(8):308-314. Publisher Full Text

  1. Edwards JS, Palsson BO: Metabolic flux balance analysis and the in silico analysis of Escherichia coli K-12 gene deletions. 

BMC Bioinformatics 2000, 1:1. PubMed Abstract | BioMed Central Full Text | PubMed Central Full Text

  1. Varma A, Palsson BO: Metabolic Flux Balancing: Basic concepts, Scientific and Practical Use. 

Nat Biotechnol 1994, 12:994-998. Publisher Full Text

  1. Segre D, Vitkup D, Church GM: Analysis of optimality in natural and perturbed metabolic networks. 

Proc Natl Acad Sci USA 2002, 99(23):15112-15117. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Baldi P, Brunak S, Chauvin Y, Andersen CA, Nielsen H: Assessing the accuracy of prediction algorithms for classification: an overview. 

Bioinformatics 2000, 16(5):412-424. PubMed Abstract | Publisher Full Text

  1. Price ND, Schellenberger J, Palsson BO: Uniform Sampling of Steady State Flux Spaces: Means to Design Experiments and to Interpret Enzymopathies. 

Biophysical Journal 2004, 87(4):2172-2186. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Price ND, Thiele I, Palsson BO: Candidate states of Helicobacter pylori’s genome-scale metabolic network upon application of “loop law” thermodynamic constraints. 

Biophysical J 2006, 90(11):3919-3928. Publisher Full Text

  1. Schuetz R, Kuepfer L, Sauer U: Systematic evaluation of objective functions for predicting intracellular fluxes in Escherichia coli. 

Mol Syst Biol 2007, 3:119. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Patil KR, Nielsen J: Uncovering transcriptional regulation of metabolism by using metabolic network topology. 

Proc Natl Acad Sci USA 2005, 102(8):2685-2689. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T: Cytoscape: a software environment for integrated models of biomolecular interaction networks. 

Genome Res 2003, 13(11):2498-2504. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Herrgard MJ, Swainston N, Dobson P, Dunn WB, Arga KY, Arvas M, Bluthgen N, Borger S, Costenoble R, Heinemann M, et al.: A consensus yeast metabolic network reconstruction obtained from a community approach to systems biology. 

Nat Biotech 2008, 26:1155-1160. Publisher Full Text

  1. Forster J, Famili I, Fu PC, Palsson BO, Nielsen J: Genome-Scale Reconstruction of the Saccharomyces cerevisiae Metabolic Network. 

Genome Research 2003, 13(2):244-253. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Reed JL, Patel TR, Chen KH, Joyce AR, Applebee MK, Herring CD, Bui OT, Knight EM, Fong SS, Palsson BO: Systems Approach to Genome Annotation: Prediction and Validation of Metabolic Functions. 

Proc Natl Acad Sci USA 2006, 103(46):17480-17484. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Nissen TL, Kielland-Brandt MC, Nielsen J, Villadsen J: Optimization of ethanol production in Saccharomyces cerevisiae by metabolic engineering of the ammonium assimilation. 

Metab Eng 2000, 2(1):69-77. PubMed Abstract | Publisher Full Text

  1. Roca C, Nielsen J, Olsson L: Metabolic engineering of ammonium assimilation in xylose-fermenting Saccharomyces cerevisiae improves ethanol production. 

Appl Environ Microbiol 2003, 69(8):4732-4736. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Hartman JL IV: Buffering of deoxyribonucleotide pool homeostasis by threonine metabolism. 

Proc Natl Acad Sci USA 2007, 104(28):11700-11705. PubMed Abstract | Publisher Full Text | PubMed Central Full Text

  1. Gelling CL, Piper MD, Hong SP, Kornfeld GD, Dawes IW: Identification of a novel one-carbon metabolism regulon in Saccharomyces cerevisiae. 

J Biol Chem 2004, 279(8):7072-7081. PubMed Abstract | Publisher Full Text

  1. Denis V, Daignan-Fornier B: Synthesis of glutamine, glycine and 10-formyl tetrahydrofolate is coregulated with purine biosynthesis in Saccharomyces cerevisiae. 

Mol Gen Genet 1998, 259(3):246-255. PubMed Abstract | Publisher Full Text

  1. Hjortmo S, Patring J, Andlid T: Growth rate and medium composition strongly affect folate content in Saccharomyces cerevisiae. 

Int J Food Microbiol 2008, 123(1–2):93-100. PubMed Abstract | Publisher Full Text

  1. Kussmann MRF, Affolter M: OMICS-driven biomarker discovery in nutrition and health. 

J Biotechnol 2006, 124(4):758-787. PubMed Abstract | Publisher Full Text

  1. Serkova NJ, Niemann CU: Pattern recognition and biomarker validation using quantitative 1H-NMR-based metabolomics. 

Expert Rev Mol Diagn 2006, 6(5):717-731. PubMed Abstract | Publisher Full Text

 

Read Full Post »

Older Posts »