Feeds:
Posts
Comments

Posts Tagged ‘Partners HealthCare’

8:30AM 11/13/2014 – 10th Annual Personalized Medicine Conference at the Harvard Medical School, Boston

Reporter: Aviva Lev-Ari, PhD, RN

 

REAL TIME Coverage of this Conference by Dr. Aviva Lev-Ari, PhD, RN – Director and Founder of LEADERS in PHARMACEUTICAL BUSINESS INTELLIGENCE, Boston http://pharmaceuticalintelligence.com

 

8:30 a.m. Harvard Business School Case Study: 23andMe

Harvard Business School Case Study: 23andMe

It is now a tradition and a unique feature of the Harvard Personalized Medicine Conference that Richard Hamermesh, a professor at Harvard Business School,  ‟teaches” a case as he does for the students at HBS. This year Professor Hamermesh has selected a couple of cases that were written about the company 23andMe. As usual, this case study will be highly interactive and educational. If you have not participated in one of these case studies at our Conference, you do not want to miss it!

23andMe – Harvard Business School Case Study

Leader:

Richard Hamermesh, D.B.A. @HarvardHBS

MBA Class of 1961 Professor of Management
Practice; Faculty Chair, HBS Healthcare Initiative, Harvard Business School

Two cases on 23andMe @23andMe

Active Group Discussion:

  • Customer very young in 20s and 30s – curiosity of sophisticated consumers
  • Personal interest in Private medical condition give illnesses in the family
  • Genotyping vs sequencing, only whole sequencing gives all variants
  • False Advertisement of the Service: on MNSBC, other
  • Do you allow your data to be used in Research vs and be sold to Pharma and be paid for sharing the data by the 23andMe
  • Price dropped from $250 to $99
  • 15 years from Now — No harm to Consumer, much data on Human Genome collected and Personal Medical Conditions improved by acting on information revealed by Genomics
  • 23andMe placed the Platform since genetic information was not otherwise available — They Pioneer with innovation on behalf of the Consumer
  • VC encourages Innovators to push the envelop – 23andMe did that with Private funding by Eugene Brin from Google
  • QUALITY OF INTERPRETATION OF GENOMICS information is key and it is improving

 

 

– See more at: http://personalizedmedicine.partners.org/Education/Personalized-Medicine-Conference/Program.aspx#sthash.qGbGZXXf.dpuf

 

@HarvardPMConf

#PMConf

@SachsAssociates

@HarvardHBS

@harvardmed

@23andMe

Read Full Post »

Personalized Cardiovascular Genetic Medicine at Partners HealthCare and Harvard Medical School

Curator: Aviva Lev-Ari, PhD, RN

UPDATED on 5/4/2015

Goes to Clinic @MGH: Clinically validated versions of Exome Sequencing and Analysis using Broad-developed methods like Hybrid Capture, the Genome Analysis Toolkit (GATK), and MuTect

http://pharmaceuticalintelligence.com/2015/05/04/goes-to-clinic-mgh-clinically-validated-versions-of-exome-sequencing-and-analysis-using-broad-developed-methods-like-hybrid-capture-the-genome-analysis-toolkit-gatk-and-mutect/

Center for Personalized Genetic Medicine, Partners HealthCare and Harvard Medical School

The Partners HealthCare Center for Personalized Genetic Medicine offers technologies and technical support for the research activities of Partners investigators. Our objective is to help investigators advance their research programs and to provide the highest quality service, technical expertise, and leading technologies for genomics research. Our goal is to broaden the access to these technologies while offering the best customer service in the most cost conscious and time efficient manner possible.

We are organized into four principal service areas:

  • sequence analysis,
  • genotyping,
  • expression analysis, and
  • bioprocessing/sample management

Our platforms include next generation sequencing with Illumina HiSeq2000 and GA ii analyzers as well as Sanger sequencing using ABI 3730 XL sequence analyzers. Targeted custom genotyping is offered using Sequenom and Illumina GoldenGate panels as well as GWAS scale projects using Illumina Infinium and DNA methylation analysis using Illumina bead arrays. Expression analysis is available with capabilities for processing total RNA on either Affymetrix or Illumina arrays.

Through services from our BioSample Services Facility (BSF) and Partners Biorepository for Medical Discovery (PBMD) teams we provide a research platform for handling samples in a standardized manner to provide consistency from sample to sample. The BSF is able to assist investigators to configure projects utilizing your own samples or coupled with the PCPGM-PBMD we are able to support the integration of cohorts of samples selected from the PBMD into analysis on our genomics platforms.

DNA Sequencing

The DNA Sequencing Group at the Partners HealthCare Center for Personalized Genetic Medicine has a strong history of producing high quality, dependable, and informative results for collaborators and clients. The DNA Sequencing Group participated in the Human Genome Project, building the STS-Based BAC map for Human Chromosome 12, and providing Chromosome 12 tiling path clones to the Baylor Human Genome Sequencing Center for sequencing.

The group sequenced 113 BACs for the Mouse Genome Project, contributing 24 megabases of finished mouse sequences to the published Mouse Genome, as well as providing draft sequences for unique strains of several bacterial genomes, including Pseudomonas aeruginosa, and Vibrio cholerae. More recently, the group participated in identifying mutations linked to numerous diseases, either in collaborations or by providing client laboratories with full service resequencing and analysis.

Services by Project Goals

Mutation Identification via Resequencing

This facility provides full-service resequencing of regions of interest in one or more genomic DNAs, including the following:

  • Discussion of the scope of the project and a cost quote
  • Identification of genes in the region of interest as needed, with the Investigator
  • Primer design using our automated system, to amplify desired regions
  • Primer ordering
  • QC of the primers on DNA standards, if required
  • PCR amplification of DNA provided by Investigator
  • PCR clean-up
  • Sequencing reactions
  • Sequencing reaction clean-up
  • Sequence application to the ABI 3730 XL Analyzer
  • Chromatograms are made available to Investigator over web (GIGPAD)
  • Data assembly and analysis using Phred Phrap and PolyPhred
  • One round of repeats and redesign if necessary
  • Report of variations found throughout sequence
  • Trouble shooting for 100% coverage if desired
Research Services
  • Fee-for-service sequencing
  • Fragment analysis / genotyping (Microsatellite Instability)
Technology Development
  • New technology testing and development
  • Collaborative Protocol development
  • Beta-test site for instrumentation and software
Clinical Diagnosis
  • Diagnostic test development
  • Sequencing for clinical diagnostics group
Genomic Sequencing Projects
  • Human
  • Rodent
  • Bacteria

http://pcpgm.partners.org/research-services/sequencing

Advancing Translational Genomics through Personalized Medicine Projects

The mission of the Partners HealthCare Center for Personalized Genetic Medicine (PCPGM) is to utilize genetics and genomics to promote and implement personalized medicine in caring for patients throughout the Partners HealthCare system and in health care nationally and globally.

The Personalized Medicine Project program was developed to support the clinical research efforts of junior Partners HealthCare investigators for translational genetics and genomic projects to advance personalized medicine.  The goal of this program is to identify biological markers that can be used as potential predictive tests.  This will be accomplished by:

  • Leveraging the Partners HealthCare Research Patient Data Registry (RPDR) and the Partners Biorepository for Medical Discovery (PBMD), centralized locations where Partners HealthCare patient data and/or samples are stored.
  • Identifying novel biological markers or new uses for existing markers.
  • Focusing on tests that could:
    • improve diagnostic sensitivity or specificity;
    • further stratify patient groups with a given diagnosis;
    • predict improved clinical outcomes; or
    • assist with selection of therapies or methods to manage disease.

http://pcpgm.partners.org/biorepository/pmprojectsrfp

Harvard Medical School Genetics Training Program

The Harvard Medical School (HMS) Genetics Training Program is one of the oldest and largest programs in the country. It was founded by Drs. John Littlefield at the Massachusetts General Hospital and Park Gerald at Children’s Hospital Boston in the early 1970’s. The program has trained scientists and clinicians who have become leaders in academic genetics, and has supported investigators who have made major contributions to the clinical practice of genetics and genetics research.

The HMS Genetics Training Program is accredited by the ABMG in all areas of training – Clinical Genetics, Biochemical Genetics, Cytogenetics, and Molecular Genetics. This provides an opportunity for our trainees to become active candidates for board certification in a discipline(s) of medical genetics in addition to receiving laboratory training. The training laboratories and clinics of the program are centered at HMS and its affiliated institutions including Brigham and Women’s Hospital (BWH), the HMS Department of Genetics, Beth Israel Deaconess Medical Center (BIDMC), Children’s Hospital Boston (CHB), Dana Farber Cancer Institute (DFCI), and Massachusetts General Hospital (MGH). The HMS Genetics Training Program provides trainees the opportunity to take advantage of the extraordinarily rich academic environment offered at HMS and its affiliated institutions as well as the greater Boston scientific community.

Cardiovascular Research Center @MGH

The Cardiovascular Research Center was founded in 1990, and occupies over 30,000 sq. ft. of laboratory space in both the Charlestown Navy Yard and the Richard B. Simches Research Building. Dr. Mark Fishman, now president of the Novartis Institutes for Biomedical Research, directed the Center from 1990 until 2002. From 2002-2005, Dr. Kenneth Bloch served as Interim Director and then in June 2005, the Massachusetts General Hospital welcomed Dr. Kenneth Chien as the new scientific director of the Cardiovascular Research Center. Prior to his MGH appointments, Dr. Chien directed the Institute for Molecular Medicine at the University of California at San Diego. An internationally recognized biologist specializing in cardiovascular science, he is a true pioneer in developing new therapeutic strategies to prevent the onset and progression of heart failure. Dr. Chien served as director until 2012.

Cardiovascular Research Center investigators have made many groundbreaking discoveries. Among these include:

• first identification of progenitor cells in the heart
• cloning of the first vertebrate cell death genes
• knocking out the genes that produce nitric oxide (NO), showing the importance of this molecule to atherosclerosis and stroke
• clinical use of NO to treat patients with pulmonary hypertension
• development of gene and cell transfer approaches to treat heart failure
• performance of the first large-scale genetic screen in a vertebrate (the zebrafish)
• identification of genes critical to cardiac pacemaking, rhythm, contractile function, and normal heart patterning
• discovery of a new methylase gene responsible for altering DNA structure during an individual’s lifetime

The Cardiovascular Research Center has taken great pride in the training of scientists with MDs and/or PhDs, as well as graduate students from a variety of Boston area institutions.

The Cardiovascular Research Center has two locations, one in the Charlestown Navy Yard and the other on the main campus’s Charles River Plaza complex in the Richard Simches Research Center.

Both the Simches and Navy Yard sites offer state of the art facilities, including tissue culture rooms, warm and cold rooms, histology rooms, autoclave facilities, hot labs, scope rooms and conference rooms. The Navy Yard lab has a topnotch zebrafish facility that is utilized by many scientists both inside and outside the Center, and a transgenic mouse core for both knock-ins and knock-outs. The Navy Yard facilities also contain echocardiogram equipment, specialized microscopes equipped with video capability for making movies, as well as a confocal microscope available to the Center researchers. The Simches lab houses the CVRC Stem Cell Biology + Therapy program, including a dedicated facility for human ES cell based technology, run by Dr. Chad Cowan, and future plans for high throughput screening facility to allow chemical screening in ESX cell based systems. Other cores available to researchers include a Cell Sorting and Flow Cytometry lab and a DNA sequencing core.

The Cardiology Laboratory for Integrative Physiology & Imaging lab is dedicated to large animal studies. An in house interventional cardiologist specializing in large animals performs the surgeries. In addition there are technicians that assist in the daily operations of the lab and can assist in experiment design and project implementation. This lab specializes in large animal imaging, CAT scans and catheter base manipulations. There is also an MRI imaging facility housed in the lab.

http://www2.massgeneral.org/cvrc/about.html

Genomics and Cardiovascular Medicine @MGH

Translational Medicine: Genomics and Proteomics @MGH

The goal of the Translational Medicine Program is to harness the rich clinical cardiovascular population at the Massachusetts General Hospital to identify and validate novel genomic determinants of cardiovascular disease. Our goal is not to capture the entire cohort of cardiovascular patients presenting to Massachusetts General Hospital, but rather to focus our efforts on extremely well-phenotyped human models that are unique to cardiovascular disease. Of particular interest are “perturbational” studies in humans (e.g., cardiac exercise testing) that elicit robust phenotypes in affected individuals to serve as the springboard for analyses that span from genomics to proteomics and biochemical profiling. The Translational Medicine Program will involve a multidisciplinary group of investigators who contribute expertise in cardiovascular basic science, clinical cardiology, genetic/genomic epidemiology, bioinformatics, imaging, pathology, as well as clinical chemistry and mass spectrometry. While the Program in Translational Medicine will be physically located at the Massachusetts General Hospital Main Campus, the effort will leverage ongoing interdisciplinary collaborations with investigators at the Framingham Heart Study, the Broad Institute of M.I.T., Harvard University, and Harvard Medical School. Our goals are to:• Identify specific unmet needs in cardiovascular biomarker and pathway discovery (e.g., genomic markers of subclinical premature coronary artery disease, serum biomarkers of myocardial ischemia)• Match cutting-edge technologies with our unique patient cohorts for “first in man” studies• Establish the infrastructure necessary to phenotype patients with the targeted condition (from plasma samples, RNA, DNA, imaging, etc.) and enroll sufficiently sized cohort(s) with the requisite power to validate novel biomarkers.• Establish scientifically high priority research projects to target for independent funding.• Ultimately, develop novel therapeutic interventions.While efforts in translational investigation are already underway, this program will identify synergies between ongoing studies and catalyze new opportunities. Several of the ongoing projects that are anticipated to serve as cornerstones of this effort include:Proteomics and Metabolomics Studies (PI: Gerszten , Wang)
Recent advances in proteomic and metabolic profiling technologies have enhanced the feasibility of high throughput patient screening for the diagnosis of disease states. Small biochemicals and proteins are the end result of the entire chain of regulatory changes that occur in response to physiological stressors, disease processes, or drug therapy. In addition to serving as biomarkers, both circulating metabolites and proteins participate as regulatory signals, such as in the control of blood pressure. Our ongoing studies have helped pioneer the application of novel mass spectrometry and liquid chromatography techniques to plasma analysis. In parallel with the profiling efforts, we have developed statistical software for functional pathway trend analysis and used it to demonstrate significant coordinate changes in specific pathways. Such analysis allows us to gain insight into the functionally relevant cellular mechanisms contributing to disease pathways and increases the likelihood that prospective biomarkers will be validated in other patient cohorts. Support for this effort would be synergistic with ongoing funding, including the recent appointment and support for Dr. Gerszten to lead a metabolomics initiative at the Broad Institute.Cardiovascular Genetics and Genomics Studies (PIs: KathiresanNewton-Cheh,Wang, and O’Donnell)
Through the Human Genome Project and the International Haplotype Map project, researchers now have available the complete human genome sequence, a nearly complete set of common single nucleotide polymorphisms (SNPs), and a map of the patterns of correlation (“linkage disequilibrium”) among SNPs. Research on a large-scale is now possible to define associations of common, complex human cardiovascular diseases —such as myocardial infarction and sudden cardiac death—with genetic variants using candidate gene and genome-wide association studies, gene sequencing, and family-based linkage studies. Specific diseases and traits being studied by CVRC researchers include early-onset myocardial infarction, sudden cardiac death, blood lipids, blood pressure, electrocardiographic QT interval and blood hemostatic factor levels. These studies draw clinical material from the Massachusetts General Hospital and from collaborations with population-based epidemiologic cohorts such as the Framingham Heart Study. Like the metabolomics/proteomics work, these efforts build on the technologic and scientific expertise at the Broad Institute. Specifically, CVRC researchers leverage the Broad Institute’s expertise in large-scale genotyping, genomics, and statistical genetics. The collaboration between the Massachusetts General Hospital, the Framingham Heart Study, and the Broad Institute brings together resources that are unique to each institution to identify genes related to complex cardiovascular traits and to ultimately impact human health.Chemical Biology Program (PIs: Peterson and Shaw) Dr. Peterson’s group has championed the zebrafish as a tool for drug discovery. The zebrafish has become a widely used model organism because of its fecundity, its morphological and physiological similarity to mammals, the existence of many genomic tools and the ease with which large, phenotype-based screens can be performed. Because of these attributes, the zebrafish also provides opportunities to accelerate the process of drug discovery. By combining the scale and throughput of in vitro screens with the physiological complexity of animal studies, the zebrafish promises to contribute to several aspects of the drug development process, including target identification, disease modeling, lead discovery and toxicology. The Program in Translational Medicine will specifically support efforts to test novel pro-angiogenic factors (discovered as suppressors of the “gridlock” phenotype in zebrafish) on human cells such as circulating endothelial precursors.Dr. Shaw’s group is studying the cellular effects of human disease mutations in patient samples, by perturbing cells with a panel of thousands of drugs, and asking whether mutant versus wild-type cells react differently to a given biochemical (reminiscent of a genetic interaction screen). Dr. Shaw has demonstrated the feasibility of this approach using lymphoblast cell lines from a family affected by a monogenic form of diabetes (MODY1), and shown that glucocorticoid signaling differs between affected vs. unaffected patients. Because his studies incorporate the use of FDA-approved drugs, he can quickly identify both potentially “druggable” disease pathways as well as novel therapeutic agents. Further validation of these efforts in other monogenic disorders, such as LDL-receptor deficient patients is planned next. Ultimately this work will be extended to studies in complex genetic diseases.Director: Rob Gerszten, MDPrincipal Investigators:
• Farouc Jaffer, MD, PhD
• Sekar Kathiresan, MD
• Chris Newton-Cheh, MD, MPH
• Randall Peterson, PhD
• Stanley Shaw, MD, PhD
• Thomas Wang, MD

Genetic Basis of Cardiomyopathy

Original gene identification for Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy, Autosomal Dominant

McNally E, MacLeod H, Dellefave L. Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy, Autosomal Dominant. 2005 Apr 18 [Updated 2009 Oct 13]. In: Pagon RA, Bird TD, Dolan CR, et al., editors. GeneReviews™ [Internet]. Seattle (WA): University of Washington, Seattle; 1993-.

Summary

Disease characteristics. Autosomal dominant arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C) is characterized by progressive fibrofatty replacement of the myocardium that predisposes to ventricular tachycardia and sudden death in young individuals and athletes. It primarily affects the right ventricle; with time, it may also involve the left ventricle. The presentation of disease is highly variable even within families, and affected individuals may not meet established clinical criteria. The mean age at diagnosis is 31 years (±13; range: 4-64 years).

Available from:

http://www.ncbi.nlm.nih.gov/books/NBK1131/

Pan Cardiomyopathy Panel

@the Center for Personalized Genetic Medicine of Partners HealthCare and Harvard Medical School

The Pan Cardiomyopathy (PCM) Panel contains 51 cardiomyopathy genes including Titin (TTN), which encodes the largest human protein. This panel covers genes associated with HCM, DCM, RCM, LVNC, ARVC and CPVT and uses a combination of Next Generation Sequencing technology and conventional Sanger sequencing.

For illustrative reference, click to see one of our images or diagrams. Genes on Pan Cardiomyopathy Panels, Disease-Gene AssociationsGene Cellular Location.

Please select on the disease to read moreHCM,DCMARVC/CPVT, or LVNC.

Current Tests:

Pan Cardiomyopathy Panel – 51 genes

  • HCM Panel – 18 genes§
  • DCM Panel – 27 genes§
  • ARVC/CPVT Panel – 8 genes§
  • LVNC Panel – 10 genes§

§Optional reflex to remaining genes

Storage Cardiomyopathy – please select a disease to learn more

For any other single gene tests, please call the LMM at 617-768-8499 or lmm@partners.org.

For Variant Classification Rules – Lab for Molecular Medicine (LMM)

http://pcpgm.partners.org/sites/default/files/LMM/Resources/LMM_VariantClassification_05.26.11.pdf 

For LMM Reference Sequences

http://pcpgm.partners.org/sites/default/files/LMM/Resources/LMMRefSeq-2.20.13.pdf

When to order which panel?

The Pan Cardiomyopathy panel may shorten the “testing odyssey” when a clear diagnosis has not been established. However, because many genes have not yet been associated with more than one cardiomyopathy, interpretation of novel variants may be more difficult when they are found in a gene that is not (yet) known to cause the patient’s cardiomyopathy. Please note: We are expecting an increase in “variants of unknown significance” and recommend careful consideration of the following factors when deciding whether to order the full panel or the disease specific sub-panels. The Pan Cardiomyopathy Panel may be best suited for patients who have already exhausted current testing options or whose clinical diagnosis is not yet clear. It may also be a good first line test for patients who have a family history where the number of living affected relatives would allow segregation analysis to establish or rule out pathogenicity for “variants of unknown significance (VUSs)”. Finally, the patient’s personal preferences should be considered as VUSs can cause anxiety.

Disease Backgrounds

Hypertrophic cardiomyopathy (HCM) is characterized by unexplained left ventricular hypertrophy (LVH) in a non-dilated ventricle. With a prevalence estimated to be ~1/500 in the general population, HCM is the most common monogenic cardiac disorder. To date, over 1000 variants have been identified in genes causative of HCM, most of which affect the sarcomere, the contractile unit of the cardiac muscle. In addition, defects in genes involved in storage diseases, such as LAMP2, PRKAG2 and GLA, typically cause systemic disease but may also result in predominant cardiac manifestations, which can mimic hypertrophic cardiomyopathy (HCM). For additional information about HCM, please visit GeneReviews. 

Dilated cardiomyopathy (DCM) is characterized by ventricular chamber enlargement and systolic dysfunction with normal left ventricular wall thickness. The estimated prevalence of DCM is 1/2,500 and about 20-35% of cases have a family history showing a predominantly autosomal mode of inheritance. To date, over 40 genes have been demonstrated to cause DCM, encoding proteins involved in the sarcomere, Z-disk, nuclear lamina, intermediate filaments and the dystrophin-associated glycoprotein complex. Variants in some genes cause additional abnormalities: LMNA variants are frequently found in DCM that occurs with progressive conduction system disease. Variants in the TAZ gene cause Barth syndrome, an X-linked cardioskeletal myopathy in infants. In addition, variants in several genes (including LMNA, DES, SGCD, TCAP and EMD) can cause DCM in conjunction with skeletal myopathy.  For additional information about DCM, please visit GeneReviews.

Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC) is estimated to affect approximately 1/5,000 individuals in the general population, about half of which have a family history. The disease is characterized by replacement of myocytes by fatty or fibrofatty tissue, mainly in the right ventricle. The resulting manifestations are broad and include ventricular tachyarrhythmias and sudden death in young individuals and athletes. ARVC is typically inherited in an autosomal dominant fashion with incomplete penetrance and variable expressivity and to date, 5 ARVC genes (DSP, DSC2, DSG2, PKP2, TMEM43) have been identified, all but one (TMEM43) encode components of the desmosome. For more information about ARVC, please visit GeneReviews.

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is typically characterized by exercise induced syncope due to ventricular tachycardia in individuals without structural heart disease. Two CPVT genes are known to date (RYR2 – autosomal dominant; CASQ2 – autosomal recessive). For more information about CPVT, please visit GeneReviews.

Left ventricular noncompaction (LVNC) has recently been established as a specific type of cardiomyopathy and is characterized by a spongy appearance of the left ventricular myocardium, resulting from an arrest in normal cardiac development. LVNC can be found in isolation or in association with other cardiomyopathies (HCM, DCM) as well as congenital cardiac abnormalities. The population prevalence is not known but LVNC is reported in ~0.014% of echocardiograms. LVNC is often familial and the genetic spectrum is beginning to emerge although it is not yet well defined. LVNC genes reported to date include ACTC, DTNA, LDB3, MYBPC3, MYH7, TAZ, and TNNT2 (Montserrat 2007, Klaassen 2008; Kaneda 2007, Zaragoza 2007; reviewed in: Maron 2006, Finsterer 2009). For more information about LVNC, please visit OMIM.org.


For any additional information, please contact us at 617-768-8500 or lmm@partners.org.

SOURCE:
Genes: 51 genesMethodology: A combination of next generation sequencing technology and Sanger sequencingAnalytical Sensitivity:Substitutions: 100% (95%CI=98.5-100)Small InDels: 95% (95%CI=83-99)Clinical Sensitivity: See below.Additional Links:

Cardiomyopathy

Price TAT CPT Codes
Pan Cardiomyopathy Panel (51 Genes)  –  lmPCM-pnlAv2_L 
$3,950 8-12 wks 81479
HCM Panel (18 Genes)  –  lmPCM-pnlB_L
$3,200 8-12 wks 81479
DCM Panel (27 Genes)  –  lmPCM-pnlCv2_L
$3,850 8-12 wks 81479
ARVC/CPVT Panel (8 Genes)  –  lmPCM-pnlD_L
$3,000 8-12 wks 81479
LVNC Panel (10 Genes)  –  lmPCM-pnlE_L
$3,200 8-12 wks 81479
Remaining Pan Cardiomyopathy Genes (HCM Reflex)  –  lmPCM-pnlFv2_L
$2,000 8-12 wks 81479
Remaining Pan Cardiomyopathy Genes (DCM Reflex)  –  lmPCM-pnlGv2_L
$2,000 8-12 wks 81479
Remaining Pan Cardiomyopathy Genes (ARVC/CPVT Reflex)  –  lmPCM-pnlHv2_L
$2,000 8-12 wks 81479
Remaining Pan Cardiomyopathy Genes (LVNC Reflex)  –  lmPCM-pnlIv2_L
$2,000 8-12 wks 81479
Remaining Pan Cardiomyopathy Genes (Version 1 Reflex) – lmPCM-pnlL_L
$750 8-12 wks 81479
Unexplained Cardiac Hypertrophy Panel (2 genes)  –  lmUCH-pnlA_L
$1,500 3 wks 81479
ABCC9 Gene Sequencing  –  lmABCC9-a_L
$1,800 3 wks 81479
ACTC Gene Sequencing  –  lmACTC-a_L
$700 3 wks 81405
ACTN2 Gene Sequencing  –  lmACTN2-a_L
$1,500 3 wks 81479
CSRP3 Gene Sequencing  –  lmCSRP3-a_L
$900 3 wks 81479
CTF1 Gene Sequencing  –  lmCTF1-a_L
$800 3 wks 81479
DES Gene Sequencing  –  lmDES-a_L
$750 3 wks 81479
DSC2 Gene Sequencing  –  lmDSC2-a_L
$1,150 3 wks 81479
DSG2 Gene Sequencing  –  lmDSG2-a_L
$1,075 3 wks 81479
DSP Gene Sequencing  –  lmDSP-a_L
$1,700 3 wks 81479
DTNA Gene Sequencing – lmDTNA-a_L
$1,500 5-6 wks 81479
EMD Gene Sequencing  –  lmEMD-a_L
$450 3 wks 81479
GLA Gene Sequencing  –  lmGLA-a_L
$700 3 wks 81405
LAMP2 Gene Sequencing  –  lmLAMP2-a_L
$700 3 wks 81405
LDB3 Gene Sequencing  –  lmLDB3-a_L
$950 3 wks 81406
LMNA Gene Sequencing  –  lmLMNA-a_L
$700 3 wks 81406
MYBPC3 Gene Sequencing  –  lmMYBPC3-a_L
$1,500 3 wks 81407
MYH7 Gene Sequencing  –  lmMYH7-a_L
$1,700 3 wks 81407
MYL2 Gene Sequencing  –  lmMYL2-a_L
$700 3 wks 81405
MYL3 Gene Sequencing  –  lmMYL3-a_L
$700 3 wks 81405
PKP2 Gene Sequencing  –  lmPKP2-a_L
$1,500 3 wks 81479
PLN Gene Sequencing  –  lmPLN-a_L
$400 3 wks 81479
PRKAG2 Gene Sequencing  –  lmPRKAG2-a_L
$1,000 3 wks 81406
SCN5A Gene Sequencing – lmSCN5A-a_L
$1,700 5-6 wks 81407
SGCD Gene Sequencing  –  lmSGCD-a_L
$1,100 3 wks 81405
TAZ Gene Sequencing  –  lmTAZ-a_L
$700 3 wks 81406
TCAP Gene Sequencing  –  lmTCAP-a_L
$700 3 wks 81479
TMEM43 Gene Sequencing  –  lmTMEM43-a_L
$700 3 wks 81479
TNNI3 Gene Sequencing  –  lmTNNI3-a_L
$700 3 wks 81405
TNNT2 Gene Sequencing  –  lmTNNT2-a_L
$1,000 3 wks 81406
TPM1 Gene Sequencing  –  lmTPM1-a_L
$700 3 wks 81405
TTN Gene Sequencing  –  lmTTN-a_L
$3,000 8-12 wks 81479
TTR Gene Sequencing – lmTTR-a_L
$485 3 wks 81404
VCL Gene Sequencing  –  lmVCL-a_L
$1,500 3 wks 81479

Congenital Heart Disease/Defects

Price TAT CPT Codes
Congenital Heart Disease Panel A (GATA4, NKX2-5, JAG1)  –  lmCHD-pnlA_L
$1,300 4 wks 81479
ELN (Elastin) Gene Sequencing  –  lmELN-a_L
$1,300 4 wks  81479
GATA4 Gene Sequencing  –  lmGATA4-a_L
$750 3 wks 81479
JAG1 Gene Sequencing  –  lmJAG1-a_L
$1,100 3 wks 81407
NKX2-5 Gene Sequencing  –  lmNKX2-5-a_L
$600 3 wks 81479
SOURCE:

Lakdawala NK, Funke BH, Baxter S, Cirino A, Roberts AE, Judge DP, Johnson N, Mendelsohn NJ, Morel C, Care M, Chung WK, Jones C, Psychogios A, Duffy ERehm HL, White E, Seidman JG, Seidman CE, Ho CY.  Genetic Testing for Dilated Cardiomyopathy in Clinical Practice. J Card Fail 2012, In press.

Neri PM, Pollard SE, Volk LA, Newmark L, Varugheese M, Baxter S, Aronson SJRehm HL, Bates DW. Usability of a Novel Clinician Interface for Genetic ResultsJ Biomed Informatics. 2012. In press.

Genomics @Brigham and Women’s Hospital and Harvard Medical School  

The goal of The Cardiovascular Genome Unit (TCGU) is to foster interdisciplinary interaction between clinical investigators and scientists to comprehensively explore the era of human genomic research. In particular, our aim would be to identify, categorize and characterize the genes and genetic pathways of the vascular and cardiac tissues of the cardiovascular system during oncogenesis, normal function and the pathogenesis of cardiovascular diseases.

    The Cardiovascular Genome Unit is responsible for indexing gene expression, profiling gene expression, identifying SNPs and generation of protein profiles from a wide variety of tissues representative of various anatomical regions as well as developmental and pathological stages in the cardiovascular system. This information resource emphasizes on cardiovascular disease and should aid in the discovery of disease causing genes, diagnostic and prognostic markers, drug targets, protein therapeutics and improved therapeutic strategies for cardiovascular disease.

    Our laboratory is the curator of a genome-based resource for molecular cardiovascular medicine consisting of over 52,000 ESTs generated from nine heart and artery libraries, representing different developmental stages and disease states (Liew et al 1994, Hwang et al 1997, Dempsey et al 2000). 

    This comprehensive catalogue of cardiac and hematopoietic genes is an unmined molecular resource for microarray analysis and a genetic gold mine for the discovery of genes that may play a role in cardiovascular disorders. In order to exploit this raw data, we propose to develop cDNA microarrays consisting of known and novel sequence-tagged genes. The arrayed clones provide an excellent substrate for expression profiling of cardiovascular disease, for example heart failure or ischemic heart disease, leading the potential discovery of diagnostic as well as prognostic markers.

    In order to accomplish the goals of the center, several cutting edge technologies are being employed.

The human cardiovascular research component of our labs.

One of the most efficient and effective strategies for the identification genes is the Expressed Sequence Tag (EST) approach.  In this approach, randomly selected cDNA clones are subjected to automated sequencing (PCR or plasmid templates) to generate a partial sequence from either the 5’- or 3’-end termed an EST.  This method allows for large-scale gene tagging and indexing from any tissue- or cell-type of interest.  A comprehensive cardiovascular gene index could be developed using a variety of cardiovascular tissues representing different anatomical, developmental and pathological states.

Comparing transcript profiles between different development or disease states is a powerful way to gain insight into the genetic changes underlying these events.  This is especially important when looking at complex systems, such as in development or disease (e.g. hypertension or atherosclerosis).  There are several unique approaches to this problem, several of which are:

a)      EST profile Comparison– After the production of a significant number of ESTs from 2 or more libraries, the frequencies of ESTs can be compared to identify those genes which are differentially expressed.     However, normalized or subtracted cDNA libraries cannot be used for this and this method is most effective for finding large differences in expression.

 

b)      cDNA Microarray Hybridization– The recent introduction of the cDNA microarray, a technology capable of analyzing the expression of thousands of genes simultaneously in a single experimentmay  provide one of the best ways to delineate gene expression patterns.  In the cDNA microarray, cDNA clones are spotted onto a glass slide matrix and hybridized with fluorescently labeled cDNA probes derived from total RNA pools of test and reference cells or tissues.  The signal intensity for each probe is quantified and any differences between the two samples becomes readily apparent.  Thus, the genetic changes underlying the phenotype of study can be identified at the level of a single gene. 

 

c)      Identification of Single Nucleotide Polymorphisms– SNPs are single-base heritable variations in the genome which occur once in approximately 1000 bases in the human genome and occur at a frequency of >1% in the human population.  SNPs provide an important genetic resource useful for disease gene discovery. including the identification of disease susceptible genes.  SNPs can be identified through comparison of EST sequences, DNA hybridization strategies and direct sequencing of genomic DNA.  The generation of a SNP database for genes expressed in the cardiovascular system will provide a valuable resource to aid in disease gene discovery. 

 

d)      Quantitative determination of expressed genes– the up- and down- regulated genes are crucial to the phenotypic expression of any given cell.  The frequency of gene expressed in development or disease state can be obtained from an EST approach using cDNA libraries as well as its intensity detected using microarrays.  Such results can be verified through RT-PCR analysis from the tissue samples.  A high through-put analysis of 96 samples can be performed by real-time PCR analyses.

Using our 10,000 element “CardioChip”, we elucidated over 100 differentially expressed genes in end-stage heart failure resulting from dilated cardiomyopathy. The results were published in

Am J Pathol. 2002 June; 160(6): 2035–2043.

Global Gene Expression Profiling of End-Stage Dilated Cardiomyopathy Using a Human Cardiovascular-Based cDNA Microarray

From Cardiovascular Genome Unit*, the Department of Medicine, and the Department of Anesthesiology,Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts; and the Department of Laboratory Medicine and Pathobiology,University of Toronto, Toronto, Ontario, Canada

Abstract

To obtain a genomic portrait of heart failure derived from end-stage dilated cardiomyopathy (DCM), we explored expression analysis using the CardioChip, a nonredundant 10,848-element human cardiovascular-based expressed sequence tag glass slide cDNA microarray constructed in-house. RNA was extracted from the left ventricular free wall of seven patients undergoing transplantation, and five nonfailing heart samples. Cy3- and Cy5-labeled (and reverse dye-labeled) cDNA probes were synthesized from individual diseased or nonfailing adult heart RNA, and hybridized to the array. More than 100 transcripts were consistently differentially expressed in DCM >1.5-fold (versus pooled nonfailing heart,P < 0.05). Atrial natriuretic peptide was found to be up-regulated in DCM (19-fold compared to nonfailing, P < 0.05), as well as numerous sarcomeric and cytoskeletal proteins (eg, cardiac troponin, tropomyosin), stress response proteins (eg, HSP 40, HSP 70), and transcription/translation regulators (eg, CCAAT box binding factor, eIF-1AY). Down-regulation was most prominently observed with cell-signaling channels and mediators, particularly those involved in Ca2+ pathways (Ca2+/calmodulin-dependent kinase, inositol 1,4,5-trisphosphate receptor, SERCA). Most intriguing was the co-expression of several novel, cardiac-enriched expressed sequence tags. Quantitative real-time reverse transcriptase-polymerase chain reaction of a selection of these clones verified expression. Our study provides a preliminary molecular profile of DCM using the largest human heart-specific cDNA microarray to date.

Dilated cardiomyopathy (DCM) is characterized clinically by left ventricular dilatation, wall thinning, and homogeneous dysfunction of the myocardium leading to congestive heart failure. Genetically, DCM seems to evolve through primary mutations in the genes of the sarcomeric proteins. 1 However, recent evidence suggests that, despite distinct pathways leading to divergent endpoint phenotypes of each disease, there may exist some overlapping genetic modifiers leading to a conversion of one to the other. 2 How this occurs is under question; to understand this, a better knowledge of the molecular pathways and intermediary regulators is required.

Global analysis of gene expression has proven to be a fruitful means of examining the overall molecular portrait of a particular event as well as seeking out novel candidate transcripts that may play a role in formulating the phenotype or genotype of interest. By using this strategy, multiple genes and pathways in complex disorders can be visualized simultaneously, allowing for a feasible platform from which to investigate new and interesting genes. Using expressed sequence tag technology, our laboratory has generated a compendium of genes expressed in the human cardiovascular system, with the ultimate goal of assembling the intricacies of development and of disease, particularly the pathways leading to heart failure. 3 Through a computer-based in silico strategy, we have been able to identify—in a large scale—both known and previously unsuspected genetic modulators contributing to the growth of the myocardium from fetal through adult, and from normal to a perturbed hypertrophic phenotype. In contrast a gene-by-gene approach in elucidating the genes and mechanisms involved is time-consuming and cumbersome.

Recently, microarray technology has been used as a means of large-scale screening of vast numbers of genes—if not whole genomes—that possess differential expression in two distinct conditions. Although new and exciting developments have arisen in such fields as cancer 4 and yeast, 5 advances in understanding the complexity of cardiovascular disease, 6 specifically DCM, have been limited. One recent study examined gene expression in two failing hearts using oligo-based arrays. 7 Although the GeneChip® (Affymetrix, Santa Clara, CA) offers a carefully controlled systematic method of analysis, its current lack of user flexibility in its design hinders novel gene discovery currently available in tissue-specific arrays. Our laboratory has taken advantage of our vast previously acquired resources and has constructed what we believe to be the first ever custom-made cardiovascular-based cDNA microarray, which we term the “CardioChip.” 8 Its practicality and flexibility has allowed us to conceptualize the molecular events surrounding end-stage heart failure.

This report describes the most informative cDNA microarray-based analysis of end-stage heart failure derived from DCM currently available. Although we believe we have effectively demonstrated reproducibility and reliability of our technology (both for the entire array and for a selection of genes located on it), a larger n from our population would enhance the validity of our conclusions. Certainly, there exists no homogeneous heart failure genotype, especially among only seven DCM patients. Nonetheless, we have demonstrated a common expression pattern among our set of samples, from both microarray and QRT-PCR analysis. We are also limited by the genes (both in number and identity) present on this array. Although we are currently unable to spot every gene and gene cluster on our CardioChip, we have tried to draw from a diverse assortment of genes and gene pathways, both known and unknown. It must be emphasized that this investigation is not exhaustive; by no means does it attempt to fully characterize the molecular basis of heart failure. Its intention is to provide a preliminary portrait of global gene expression in complex cardiovascular disease using cDNA microarray and QRT-PCR technology, and to highlight the effectiveness of our ever-evolving platform for gene discovery. With even more patient samples and a CardioChip toward completeness, we will be in a better position to reap the important benefits from this initial work and expand our body of knowledge.

REFERENCES

http://www.pnas.org/content/91/22/10645.full.pdf

1. Towbin JA, Bowles NE: Genetic abnormalities responsible for dilated cardiomyopathy. Curr Cardiol Rep 2000, 2:475-480. [PubMed]
2. Chien KR: Stress pathways and heart failure. Cell 1999, 98:555-558. [PubMed]
3. Hwang JJ, Dzau VJ, Liew CC: Genomics and the pathophysiology of heart failure. Curr Cardiol Rep2001, 3:198-207. [PubMed]
4. Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Tran T, Yu X, Powell JI, Yang L, Marti GE, Moore T, Hudson J, Jr, Lu L, Lewis DB, Tibshirani R, Sherlock G, Chan WC, Greiner TC, Weisenburger DD, Armitage JO, Warnke R, Levy R, Wilson W, Grever M, Byrd JC, Botstein D, Brown PO, Staudt LM: Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 2000, 403:503-511. [PubMed]
5. Gasch AP, Spellman PT, Kao CM, Carmel-Harel O, Eisen MB, Storz G, Botstein D, Brown PO: Genomic expression programs in the response of yeast cells to environmental changes. Mol Biol Cell 2000, 11:4241-4257. [PMC free article] [PubMed]
6. Taylor LA, Carthy CM, Yang D, Saad K, Wong D, Schreiner G, Stanton LW, McManus BM: Host gene regulation during coxsackievirus B3 infection in mice: assessment by microarrays. Circ Res 2000, 87:328-334. [PubMed]
7. Yang J, Moravec CS, Sussman MA, DiPaola NR, Fu D, Hawthorn L, Mitchell CA, Young JB, Francis GS, McCarthy PM, Bond M: Decreased SLIM1 expression and increased gelsolin expression in failing human hearts measured by high-density oligonucleotide arrays. Circulation 2000, 102:3046-3052.[PubMed]
8. Barrans JD, Stamatiou D, Liew CC: Construction of a human cardiovascular cDNA microarray: portrait of the failing heart. Biochem Biophys Res Commun 2001, 280:964-969. [PubMed]
9. Hwang DM, Hwang WS, Liew CC: Single pass sequencing of a unidirectional human fetal heart cDNA library to discover novel genes of the cardiovascular system. J Mol Cell Cardiol 1994, 26:1329-1333.[PubMed]
10. Hwang DM, Dempsey AA, Lee CY, Liew CC: Identification of differentially expressed genes in cardiac hypertrophy by analysis of expressed sequence tags. Genomics 2000, 66:1-14. [PubMed]
11. Hwang DM, Dempsey AA, Wang RX, Rezvani M, Barrans JD, Dai KS, Wang HY, Ma H, Cukerman E, Liu YQ, Gu JR, Zhang JH, Tsui SK, Waye MM, Fung KP, Lee CY, Liew CC: A genome-based resource for molecular cardiovascular medicine: toward a compendium of cardiovascular genes. Circulation 1997,96:4146-4203. [PubMed]
12. Liew CC, Hwang DM, Fung YW, Laurenssen C, Cukerman E, Tsui S, Lee CY: A catalogue of genes in the cardiovascular system as identified by expressed sequence tags. Proc Natl Acad Sci USA 1994,91:10645-10649. [PMC free article] [PubMed]
13. Liew CC, Hwang DM, Wang RX, Ng SH, Dempsey A, Wen DH, Ma H, Cukerman E, Zhao XG, Liu YQ, Qiu XK, Zhou XM, Gu JR, Tsui S, Fung KP, Waye MM, Lee CY: Construction of a human heart cDNA library and identification of cardiovascular based genes (CVBest). Mol Cell Biochem 1997, 172:81-87.[PubMed]
14. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ: Basic local alignment search tool. J Mol Biol1990, 215:403-410. [PubMed]
15. DeRisi J, Penland L, Brown PO, Bittner ML, Meltzer PS, Ray M, Chen Y, Su YA, Trent JM: Use of a cDNA microarray to analyze gene expression patterns in human cancer. Nat Genet 1996, 14:457-460.[PubMed]
16. Vikstrom KL, Bohlmeyer T, Factor SM, Leinwand LA: Hypertrophy, pathology, and molecular markers of cardiac pathogenesis. Circ Res 1998, 82:773-778. [PubMed]
17. Towbin JA: The role of cytoskeletal proteins in cardiomyopathies. Curr Opin Cell Biol 1998, 10:131-139.[PubMed]
18. Elliott K, Watkins H, Redwood CS: Altered regulatory properties of human cardiac troponin I mutants that cause hypertrophic cardiomyopathy. J Biol Chem 2000, 275:22069-22074. [PubMed]
19. Kimura A, Harada H, Park JE, Nishi H, Satoh M, Takahashi M, Hiroi S, Sasaoka T, Ohbuchi N, Nakamura T, Koyanagi T, Hwang TH, Choo JA, Chung KS, Hasegawa A, Nagai R, Okazaki O, Nakamura H, Matsuzaki M, Sakamoto T, Toshima H, Koga Y, Imaizumi T, Sasazuki T: Mutations in the cardiac troponin I gene associated with hypertrophic cardiomyopathy. Nat Genet 1997, 16:379-382. [PubMed]
20. Redwood C, Lohmann K, Bing W, Esposito GM, Elliott K, Abdulrazzak H, Knott A, Purcell I, Marston S, Watkins H: Investigation of a truncated cardiac troponin T that causes familial hypertrophic cardiomyopathy: Ca(2+) regulatory properties of reconstituted thin filaments depend on the ratio of mutant to wild-type protein. Circ Res 2000, 86:1146-1152. [PubMed]
21. Geisterfer-Lowrance AA, Kass S, Tanigawa G, Vosberg HP, McKenna W, Seidman CE, Seidman JG: A molecular basis for familial hypertrophic cardiomyopathy: a beta cardiac myosin heavy chain gene missense mutation. Cell 1990, 62:999-1006. [PubMed]
22. Thierfelder L, Watkins H, MacRae C, Lamas R, McKenna W, Vosberg HP, Seidman JG, Seidman CE: Alpha-tropomyosin and cardiac troponin T mutations cause familial hypertrophic cardiomyopathy: a disease of the sarcomere. Cell 1994, 77:701-712. [PubMed]
23. Arber S, Hunter JJ, Ross J, Jr, Hongo M, Sansig G, Borg J, Perriard JC, Chien KR, Caroni P: MLP-deficient mice exhibit a disruption of cardiac cytoarchitectural organization, dilated cardiomyopathy, and heart failure. Cell 1997, 88:393-403. [PubMed]
24. Dalakas MC, Park KY, Semino-Mora C, Lee HS, Sivakumar K, Goldfarb LG: Desmin myopathy, a skeletal myopathy with cardiomyopathy caused by mutations in the desmin gene. N Engl J Med 2000,342:770-780. [PubMed]
25. Satoh M, Takahashi M, Sakamoto T, Hiroe M, Marumo F, Kimura A: Structural analysis of the titin gene in hypertrophic cardiomyopathy: identification of a novel disease gene. Biochem Biophys Res Commun 1999, 262:411-417. [PubMed]
26. Olson TM, Michels VV, Thibodeau SN, Tai YS, Keating MT: Actin mutations in dilated cardiomyopathy, a heritable form of heart failure. Science 1998, 280:750-752. [PubMed]
27. Schonberger J, Seidman CE: Many roads lead to a broken heart: the genetics of dilated cardiomyopathy. Am J Hum Genet 2001, 69:249-260. [PMC free article] [PubMed]
28. Dempsey AA, Ton C, Liew CC: A cardiovascular EST repertoire: progress and promise for understanding cardiovascular disease. Mol Med Today 2000, 6:231-237. [PubMed]
29. Francis GS: Changing the remodeling process in heart failure: basic mechanisms and laboratory results. Curr Opin Cardiol 1998, 13:156-161. [PubMed]
30. Rao VU, Spinale FG: Controlling myocardial matrix remodeling: implications for heart failure.Cardiol Rev 1999, 7:136-143. [PubMed]
31. McKinsey TA, Olson EN: Cardiac hypertrophy: sorting out the circuitry. Curr Opin Genet Dev 1999,9:267-274. [PubMed]
32. Chien KR: Genomic circuits and the integrative biology of cardiac diseases. Nature 2000, 407:227-232. [PubMed]
33. Miyamoto MI, del Monte F, Schmidt U, DiSalvo TS, Kang ZB, Matsui T, Guerrero JL, Gwathmey JK, Rosenzweig A, Hajjar RJ: Adenoviral gene transfer of SERCA2a improves left ventricular function in aortic-banded rats in transition to heart failure. Proc Natl Acad Sci USA 2000, 97:793-798.[PMC free article] [PubMed]
34. Tada M, Yabuki M, Toyofuku T: Molecular regulation of phospholamban function and gene expression. Ann NY Acad Sci 1998, 853:116-129. [PubMed]
35. Marks AR: Cardiac intracellular calcium release channels: role in heart failure. Circ Res 2000, 87:8-11. [PubMed]
36. Dewaste V, Pouillon V, Moreau C, Shears S, Takazawa K, Erneux C: Cloning and expression of a cDNA encoding human inositol 1,4,5-trisphosphate 3-kinase C. Biochem J 2000, 352:343-351.[PMC free article] [PubMed]
Books (partial list):

Liew CC, Jackowski, G, Ma T, Jung, YC, Sole, MJ. Possible role of nonhistone chromatin proteins associated with heterogeneous nuclear RNA in myocardial differentiation and in the genesis of cardiomyopathy. In: Alpert NR, editor. Perspectives in
Cardiovascular Research. Raven Press; 1983. p. 497-511.

Liew CC, Takihara KY, Jandreski M, Liew J, Sole MJ. Structure and expression of human b-myosin heavy chain gene. In: Carraro U, editor. Sarcomeric and Non-sarcomeric Muscles: Basic and Applied Research Prospects for the 90s. Padova, Italy: Unipress
Padova; 1988. P 11-17.

Liew CC, Takihara KY, Liew J, Sole MJ. Characterization of human cardiac myosin heavy chain genes. In: Wu F, Wu CW, editors. Structure and Function of Nucleic Acids and Proteins, New York, Raven Press; 1990. pp.303-309.

Wang RX, Cukerman E, Chen B, Liew CC. Differential screening and megasequencing of human heart cDNA library: A search for genes associated with heart failure. In: Dhalla NS, Pierce GN, Panagia V, Beamish RE, editors. Boston: Kluwer Academic Press; 1995. P. 67-77.

Dempsey A, Liew CC. Genes involved in normal cardiac development. In: Sheridan DJ, editor. Left Ventricular Hypertrophy. London: Churchill Communications Europe Ltd; 1998: p. 61-70.

Tan K, Dempsey A, Liew CC. Cardiac genes and gene databases for cardiovascular disease genetics. In: Hollenberg NK, editor. Current Hypertension Reports. Philadelphia: Current Science Group; 1999: Vol 1:51-58.

Liew, CC. Expressed Sequence Tags. In: Encyclopedia of Molecular Medicine, Ed: T. Creighton, John Wiley and Son, New York. 2001

Hwang J-J, Dzau V and Liew CC. Genomics and thePathophysiology of Heart Failure. In: Current Cardiology Reports; Current Science Inc; 2001: Vol 3: 198-207.

Read Full Post »