Autobiography of Editorial Board Members Open Access
Copyright ©2011 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Cardiol. Apr 26, 2011; 3(4): 121-126
Published online Apr 26, 2011. doi: 10.4330/wjc.v3.i4.121
Molecular biology of heart disease
Robert Roberts, Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, K1Y 4W7, Canada
Author contributions: Roberts R solely contributed to this manuscript.
Supported by NHLBI, American Heart Association, Canadian Institutes of Health Research, Canadian Foundation for Innovation, Heart and Stroke Ontario
Correspondence to: Robert Roberts, MD, FRCPC, MACC, President and CEO, Professor of Medicine and Director, Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, K1Y 4W7, Canada. rroberts@ottawaheart.ca
Telephone: +1-613-7614779 Fax: +1-613-7614650
Received: September 13, 2010
Revised: April 13, 2011
Accepted: April 20, 2011
Published online: April 26, 2011

Abstract

Dr. Robert Roberts is currently Professor of Medicine and Director of the Ruddy Canadian Cardiovascular Genetics Centre along with being President and CEO of the University of Ottawa Heart Institute. Prior to this appointment, he was Chief of Cardiology for 23 years at Baylor College of Medicine, Houston, Texas. His original research was in cardiac enzymology which led to the development of the MBCK test which was the standard diagnostic assay for myocardial infarction for more than 3 decades. In the late 1970s, his research interests switched to molecular biology and the genetics of cardiomyopathies. He is regarded as one of the founders of molecular cardiology and has identified and sequenced more than 20 genes responsible for cardiovascular disorders. In the past 6 years, he has pursued genome-wide association studies to identify genes predisposing to coronary artery disease (CAD) and myocardial infarction. The first genetic variant for CAD, 9p21, was identified by Dr. Robert’s laboratory and, in collaboration with the international consortium, CARDIoGRAM, has identified 13 novel genes for CAD.

Key Words: Molecular biology, Genetics, Heart disease, Genome wide association studies, Genetic linkage, Creatine kinase



INTRODUCTION AND EDUCATIONAL EXPERIENCE

Robert Roberts (Figure 1) received his MD from Dalhousie University and completed his residency in Internal Medicine and Fellowship in Cardiology at the University of Toronto. Funded by a Canadian Heart Foundation Scholarship he pursued research in enzymology and cardiac metabolism at the University of California, San Diego, following which he was Director of the Cardiac Care Unit at Barnes Hospital and Associate Professor of Medicine, Washington University. In 1982, he accepted a position as Chief of Cardiology at Baylor College of Medicine and became Professor of Medicine with joint appointments in the departments of Cell Biology and Molecular Physiology and Biophysics. On April 1, 2004, Dr. Roberts was appointed President and CEO of the University of Ottawa Heart Institute and Director of The Ruddy Canadian Cardiovascular Genetics Centre. He is also an adjunct Professor of Medicine at Baylor College of Medicine.

Figure 1
Figure 1 Robert Roberts, MD, FRCPC, MACC, President and CEO, Professor of Medicine and Director, Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, K1Y 4W7, Canada.
ACADEMIC STRATEGIES AND GOALS

Dr. Roberts, in addition to his role as clinician, educator and academic leader has been a very productive scientist. His early research focused on quantification and diagnosis of ischemic heart disease. He developed the first quantitative assay for the plasma MB isoenzyme of creatine kinase (MBCK) in 1974[1,2] and the first radioimmunoassay (RIA) for MBCK[3], based on an antibody to the B-subunit in 1976, which was also the first RIA for an isoenzyme. MBCK remained the standard for the diagnosis of myocardial infarction throughout the world for more than three decades[4-12]. He was the first to purify mitochrondrial CK[13,14] and clone the cytosolic CK genes[7,15]. Today all markers for myocardial infarction, including the troponins, are antibody-based. He isolated and purified the plasma MM and MB CK subforms[8,16], elucidated the mechanism responsible for their generation, and utilized them to develop an assay for the early diagnosis of infarction[8]. His laboratory played a pivotal role in the quantification of the extent of damage associated with myocardial infarction[4,6,14] and the effect of therapies on experimental infarction[17-19] in clinical trials, including β blockers[20] and thrombolytic therapy[20-27]. Notably, the Diltiazen on Non-Q-wave Infarction Study was directed by Dr. Roberts and showed diltiazen to be an effective therapy for non-Q-wave infarction which remains the mainstay of therapy 25 years later[28].

On moving to Baylor, Dr. Robert’s basic research effort focused on the application of the techniques of recombinant DNA to cardiac growth[19,29-32] and molecular genetics. These efforts would subsequently earn him the title of one of the founders of molecular cardiology. He edited and co-authored the first textbook on Molecular Basis of Cardiology in 1993[33], and continues to author the section on Molecular Cardiology in numerous text books including Hurst’s The Heart for the past two decades[34-36]. In the early 1980s, he cloned the genes for all three human creatine kinases[7]. His achievements were sufficiently recognized by the mid-1980s, that he was chosen by the American Heart Association to direct one of the three initial Bugher Training Programs for molecular biology of the cardiovascular system. Dr. Roberts’ research has since been devoted to molecular genetics of cardiovascular disease.

ACADEMIC ACHIEVEMENTS

He has made many contributions in the field of molecular genetics on hypertrophic cardiomyopathy[37-48], familial dilated cardiomyopathy[49,50], muscular dystrophies[51,52], atrial fibrillation[45,53-56], Wolf Parkinson White Syndrome[57,58], Human eHAND[59], and arrhythmogenic right ventricular cardiomyopathy[60-62], and accomplished the following: (1) mapped the first locus for familial dilated cardiomyopathy; (2) mapped the first locus for atrial fibrillation; (3) mapped the first locus for arrhythmogenic right ventricular dysplasia in North America; (4) cloned and sequenced the desmin gene responsible for familial dilated cardiomyopathy; (5) identified the first gene for Wolff-Parkinson-White syndrome; (6) identified the troponin T mutation responsible for dilated cardiomyopathy; and (7) identified a novel family of proteins that bind specifically to triplet repeats and are responsible for myotonin mRNA nucleocytoplasmic transport[51,63-65]. He developed the only transgenic rabbit[66] with a phenotype of hypertrophic cardiomyopathy and together with transgenic mice has elucidated the pathogenesis of familial hypertrophic cardiomyopathy. Utilizing these transgenic animal models, he and his colleagues identified that statins, angiotensin II blockers and aldosterone inhibitors could reverse the phenotype[44,67,68]. In 2005, he showed that the hypertrophic cardiomyopathy phenotype in the transgenic rabbit could be prevented with atorvastatin therapy[36]. The pioneering application of genetics in research and clinical management of cardiomyopathies developed Baylor Cardiology into a major referral center for inherited cardiovascular disease.

On moving to the University of Ottawa Heart Institute, he founded The Ruddy Canadian Cardiovascular Genetics Centre. This was initiated by a $5 million donation from John and Jennifer Ruddy followed by two endowed Fellowships of $1 million each by Doug Arand and Michael Potter and Family. While his research up to this time had been on single gene disorders he now focused on genetics of common cardiovascular disorders, namely coronary artery disease (CAD). The Ottawa Heart Genomics Study was initiated in 2004 in pursuit of genes responsible for CAD and myocardial infarction. It was the first genome-wide association study (GWAS) to utilize the 500 000 DNA chip to genotype for CAD. This led to the mapping of the first locus 9p21 for CAD[69]. The risk imparted by this locus is independent of known risk factors for CAD and was published in Science on May 3, 2007[70]. The Ruddy Canadian Cardiovascular Genetics Centre, under the direction of Dr. Roberts, rapidly acquired an international reputation and the capacity to perform high throughput genotyping (> 300 million genotypes per day) and DNA sequencing. In recognition of his scientific contributions he became a member of the International Consortium, CARDIoGRAM[71], which subsequently led to the discovery of over 95 genetic risk variants regulating lipids[72], and most recently a landmark study of over 23 genetic variants with increased risk for CAD and myocardial infarction[73]. These studies have led to numerous investigations regarding the mechanism of action of 9p21, including studies in Dr. Roberts’ laboratory[74-77]. Utilizing the 9p21 gene, studies were performed which showed it could predict the severity and progression of CAD[77-79]. In a collaborative genome-wide study, his lab recently identified the first gene in the ABO group that predisposes to myocardial infarction, and ADAMTS7 which predisposes to coronary atherosclerosis without infarction[80]. He is currently involved with a GWAS to map genes predisposing to hypertension[81]. In recognition of this effort, Dr. Roberts as Principal Investigator along with his co-investigators were awarded a $12 million grant for genetic research by the Canadian Foundation for Innovation in 2006 and another 5-year grant from the Canadian Institutes of Health Research.

Dr. Roberts is world renowned as an educator, particularly in bringing the techniques of molecular biology and genetics to the cardiovascular community. He has supported this mission through several venues including national and international academic and government committees. Dr. Roberts is currently a member of the Medical Advisory Committee to the Leducq Foundation (2010-2013) and the Gairdner Foundation (2010-2013). He is also on the Board of Directors for Fields Institute. He serves on the Grant Review Committees for the Canadian Institute for Health Research (CIHR), Genome Canada, Genome Quebec, the National Heart, Lung and Blood Institute (NHLBI) and the Heart and Stroke Foundation of Ontario. Dr. Roberts chairs the Safety Monitoring of the RAMICAT clinical trial. He is the editor of Current Opinion in Cardiology and is on the editorial board of several journals. Dr. Roberts served on the Cardiovascular Study Section of the National Institutes of Health (1979-1982), the Cardiology Advisory Committee of the NHLBI (1984-1988) and subsequently the Advisory Council of the NHLBI (2000-2001). He was Chairman of the Study Section for the Cardiovascular Physiology and Pathophysiology Committee of the American Heart Association (1990-1993) and a member of the Central Research Review Committee (1990-1995). He served on the American Heart Association (AHA) Scientific Sessions Committee from 1986-1990. He became a member of the Research Planning Evaluation Committee for the AHA (1994-2001), and served as Vice-Chairman (1997-1999) and Chairman (1999-2001), and during this time, he also served on the Board of Directors for the AHA. He served as Vice President of the AHA (2001-2002). In 1991, he served as Chairman of the Scientific Sessions for the American College of Cardiology (ACC) and served on the Board of Trustees (1996-2001), Young Investigators’ Awards Committee (1988-1990), Member of Budget, Finance and Investment Committee (1997-2003), Nominating Committee (1998-2000) and Chairman of the Advisory Committee for Merck/Pfizer/ACC Foundation (2000-2006), and Member, CIHR Team Grant A (2007-Present). Dr. Roberts has lectured throughout the world and has been the plenary speaker at many national meetings including the American College of Chest Physicians Simon Rodbard Lecturer, 61st Annual Scientific Meeting of the Japanese Circulation Society, Mikamo Lecturer, Tokyo, Japan 1997, opening Plenary Speaker for the Japanese College of Cardiology 1995, Japanese Cardiology, the Secondary International Symposium on Heart Failure in Geneva, Switzerland, Simon Dack Presidential Address at the ACC Scientific Sessions (2002) and the State-of-the-Art Lecture, Canadian Cardiovascular Society (2005).

In recognition of his contributions, he has received several national and international awards. Dr. Roberts received the Distinguished Scientist Award from the ACC in 1998, the Award of Meritorious Achievement from The American Heart Association (2001), Master of the ACC (2007), and recently was awarded the McLaughlin Award from the Royal Society of Canada (2008). He was awarded the Robert Beamish Leadership Award in 2005. He has over 800 publications including Associate Editor of Hurst’s The Heart, (1989-present) and was awarded the Most Highly Cited Researcher (2002).

CONCLUSION

Dr. Roberts is a major national and international educator for molecular genetics throughout the cardiac community. He chaired and participated in a core curriculum course of molecular biology for the clinician at the AHA and ACC Annual Scientific Sessions each year for over 15 years. He has participated in the fellowship program sponsored by AHA, ACC and NHLBI annually for over 20 years. As the Director of the Bugher and NHLBI training programs, he trained more than 40 molecular cardiologists, held leadership positions in the AHA and ACC and has been recognized as an important leader in the research and practice of cardiology worldwide. Several of his fellows are Chiefs of Cardiology in the USA, Canada, Japan and several other countries.

Footnotes

Peer reviewer: Yucheng Yao, MD, Assistant Researcher, David Geffen School of Medicine, Division of Cardiology, 10833 Le Conte Ave, BH-307 CHS, Los Angeles, CA 90095-1679, United States

S- Editor Cheng JX L- Editor Cant MR E- Editor Zheng XM

References
1.  Roberts R, Henry PD, Witteeveen SA, Sobel BE. Quantification of serum creatine phosphokinase isoenzyme activity. Am J Cardiol. 1974;33:650-654.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Roberts R, Henry PD, Sobel BE. An improved basis for enzymatic estimation of infarct size. Circulation. 1975;52:743-754.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Roberts R, Sobel BE, Parker CW. Radioimmunoassay for creatine kinase isoenzymes. Science. 1976;194:855-857.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Sobel BE, Markham J, Roberts R. Factors influencing enzymatic estimates of infarct size. Am J Cardiol. 1977;39:130-132.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Roberts R, Grace AM. Purification of mitochondrial creatine kinase. Biochemical and immunological characterization. J Biol Chem. 1980;255:2870-2877.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Ritter CS, Mumm SR, Roberts R. Improved radioimmunoassay for creatine kinase isoenzymes in plasma. Clin Chem. 1981;27:1878-1887.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Perryman MB, Kerner SA, Bohlmeyer TJ, Roberts R. Isolation and sequence analysis of a full-length cDNA for human M creatine kinase. Biochem Biophys Res Commun. 1986;140:981-989.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Puleo PR, Guadagno PA, Roberts R, Perryman MB. Sensitive, rapid assay of subforms of creatine kinase MB in plasma. Clin Chem. 1989;35:1452-1455.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Ma TS, Ifegwu J, Watts L, Siciliano MJ, Roberts R, Perryman MB. Serial Alu sequence transposition interrupting a human B creatine kinase pseudogene. Genomics. 1991;10:390-399.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Friedman DL, Roberts R. Purification and localization of brain-type creatine kinase in sodium chloride transporting epithelia of the spiny dogfish, Squalus acanthias. J Biol Chem. 1992;267:4270-4276.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Ishikawa Y, Saffitz JE, Mealman TL, Grace AM, Roberts R. Reversible myocardial ischemic injury is not associated with increased creatine kinase activity in plasma. Clin Chem. 1997;43:467-475.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Puleo PR, Meyer D, Wathen C, Tawa CB, Wheeler S, Hamburg RJ, Ali N, Obermueller SD, Triana JF, Zimmerman JL. Use of a rapid assay of subforms of creatine kinase-MB to diagnose or rule out acute myocardial infarction. N Engl J Med. 1994;331:561-566.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Grace AM, Perryman MB, Roberts R. Purification and characterization of human mitochondrial creatine kinase. A single enzyme form. J Biol Chem. 1983;258:15346-15354.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Basson CT, Grace AM, Roberts R. Enzyme kinetics of a highly purified mitochondrial creatine kinase in comparison with cytosolic forms. Mol Cell Biochem. 1985;67:151-159.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Perryman MB, Strauss AW, Olson J, Roberts R. In vitro translation of canine mitochondrial creatine kinase messenger RNA. Biochem Biophys Res Commun. 1983;110:967-972.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  George S, Ishikawa Y, Perryman MB, Roberts R. Purification and characterization of naturally occurring and in vitro induced multiple forms of MM creatine kinase. J Biol Chem. 1984;259:2667-2674.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Bolli R, Myers ML, Zhu WX, Roberts R. Disparity of reperfusion arrhythmias after reversible myocardial ischemia in open chest and conscious dogs. J Am Coll Cardiol. 1986;7:1047-1056.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Myers ML, Bolli R, Lekich RF, Hartley CJ, Roberts R. N-2-mercaptopropionylglycine improves recovery of myocardial function after reversible regional ischemia. J Am Coll Cardiol. 1986;8:1161-1168.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Kugiyama K, Kerns SA, Morrisett JD, Roberts R, Henry PD. Impairment of endothelium-dependent arterial relaxation by lysolecithin in modified low-density lipoproteins. Nature. 1990;344:160-162.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Roberts R, Croft C, Gold HK, Hartwell TD, Jaffe AS, Muller JE, Mullin SM, Parker C, Passamani ER, Poole WK. Effect of propranolol on myocardial-infarct size in a randomized blinded multicenter trial. N Engl J Med. 1984;311:218-225.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  The Thrombolysis in Myocardial Infarction (TIMI) trial. Phase I findings. TIMI Study Group. N Engl J Med. 1985;312:932-936.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Turi ZG, Rutherford JD, Roberts R, Muller JE, Jaffe AS, Rude RE, Parker C, Raabe DS, Stone PH, Hartwell TD. Electrocardiographic, enzymatic and scintigraphic criteria of acute myocardial infarction as determined from study of 726 patients (A MILIS Study). Am J Cardiol. 1985;55:1463-1468.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Williams DO, Borer J, Braunwald E, Chesebro JH, Cohen LS, Dalen J, Dodge HT, Francis CK, Knatterud G, Ludbrook P. Intravenous recombinant tissue-type plasminogen activator in patients with acute myocardial infarction: a report from the NHLBI thrombolysis in myocardial infarction trial. Circulation. 1986;73:338-346.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Hsia J, Hamilton WP, Kleiman N, Roberts R, Chaitman BR, Ross AM. A comparison between heparin and low-dose aspirin as adjunctive therapy with tissue plasminogen activator for acute myocardial infarction. Heparin-Aspirin Reperfusion Trial (HART) Investigators. N Engl J Med. 1990;323:1433-1437.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Roberts R, Rogers WJ, Mueller HS, Lambrew CT, Diver DJ, Smith HC, Willerson JT, Knatterud GL, Forman S, Passamani E. Immediate versus deferred beta-blockade following thrombolytic therapy in patients with acute myocardial infarction. Results of the Thrombolysis in Myocardial Infarction (TIMI) II-B Study. Circulation. 1991;83:422-437.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Aguirre FV, McMahon RP, Mueller H, Kleiman NS, Kern MJ, Desvigne-Nickens P, Hamilton WP, Chaitman BR. Impact of age on clinical outcome and postlytic management strategies in patients treated with intravenous thrombolytic therapy. Results from the TIMI II Study. TIMI II Investigators. Circulation. 1994;90:78-86.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Sloan MA, Price TR, Petito CK, Randall AM, Solomon RE, Terrin ML, Gore J, Collen D, Kleiman N, Feit F. Clinical features and pathogenesis of intracerebral hemorrhage after rt-PA and heparin therapy for acute myocardial infarction: the Thrombolysis in Myocardial Infarction (TIMI) II Pilot and Randomized Clinical Trial combined experience. Neurology. 1995;45:649-658.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Gibson RS, Boden WE, Theroux P, Strauss HD, Pratt CM, Gheorghiade M, Capone RJ, Crawford MH, Schlant RC, Kleiger RE. Diltiazem and reinfarction in patients with non-Q-wave myocardial infarction. Results of a double-blind, randomized, multicenter trial. N Engl J Med. 1986;315:423-429.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Schneider MD, Payne PA, Ueno H, Perryman MB, Roberts R. Dissociated expression of c-myc and a fos-related competence gene during cardiac myogenesis. Mol Cell Biol. 1986;6:4140-4143.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Schneider MD, Perryman MB, Payne PA, Spizz G, Roberts R, Olson EN. Autonomous expression of c-myc in BC3H1 cells partially inhibits but does not prevent myogenic differentiation. Mol Cell Biol. 1987;7:1973-1977.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Cowley AW Jr. The Banbury Conference. Genomics to physiology and beyond: how do we get there? Physiologist. 1997;40:205-211.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Li D, Roberts R. WD-repeat proteins: structure characteristics, biological function, and their involvement in human diseases. Cell Mol Life Sci. 2001;58:2085-2097.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Roberts R Molecular basis of cardiology. Hamden, CT: Blackwell Scientific Publications 1992; .  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Roberts R, McNally E. Genetic basis for cardiovascular disease. Hurst's the heart. 13th ed. New York, NY: McGraw-Hill Inc 2009; 195-205.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Marian AJ, Brugada R, Roberts R. Cardiovascular diseases caused genetic abnormalities. Hurst's the heart. 13th ed. New York, NY: McGraw-Hill Inc 2009; 195-205.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Senthil V, Chen SN, Tsybouleva N, Halder T, Nagueh SF, Willerson JT, Roberts R, Marian AJ. Prevention of cardiac hypertrophy by atorvastatin in a transgenic rabbit model of human hypertrophic cardiomyopathy. Circ Res. 2005;97:285-292.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Perryman MB, Yu QT, Marian AJ, Mares A Jr, Czernuszewicz G, Ifegwu J, Hill R, Roberts R. Expression of a missense mutation in the messenger RNA for beta-myosin heavy chain in myocardial tissue in hypertrophic cardiomyopathy. J Clin Invest. 1992;90:271-277.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Vybiral T, Winkelmann JC, Roberts R, Joe E, Casey DL, Williams JK, Epstein HF. Human cardiac and skeletal muscle spectrins: differential expression and localization. Cell Motil Cytoskeleton. 1992;21:293-304.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Anan R, Greve G, Thierfelder L, Watkins H, McKenna WJ, Solomon S, Vecchio C, Shono H, Nakao S, Tanaka H. Prognostic implications of novel beta cardiac myosin heavy chain gene mutations that cause familial hypertrophic cardiomyopathy. J Clin Invest. 1994;93:280-285.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Greve G, Bachinski L, Friedman DL, Czernuzewicz G, Anan R, Towbin J, Seidman CE, Roberts R. Isolation of a de novo mutant myocardial beta MHC protein in a pedigree with hypertrophic cardiomyopathy. Hum Mol Genet. 1994;3:2073-2075.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Marian AJ, Zhao G, Seta Y, Roberts R, Yu QT. Expression of a mutant (Arg92Gln) human cardiac troponin T, known to cause hypertrophic cardiomyopathy, impairs adult cardiac myocyte contractility. Circ Res. 1997;81:76-85.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Oberst L, Zhao G, Park JT, Brugada R, Michael LH, Entman ML, Roberts R, Marian AJ. Dominant-negative effect of a mutant cardiac troponin T on cardiac structure and function in transgenic mice. J Clin Invest. 1998;102:1498-1505.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Karibe A, Tobacman LS, Strand J, Butters C, Back N, Bachinski LL, Arai AE, Ortiz A, Roberts R, Homsher E. Hypertrophic cardiomyopathy caused by a novel alpha-tropomyosin mutation (V95A) is associated with mild cardiac phenotype, abnormal calcium binding to troponin, abnormal myosin cycling, and poor prognosis. Circulation. 2001;103:65-71.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Lim DS, Lutucuta S, Bachireddy P, Youker K, Evans A, Entman M, Roberts R, Marian AJ. Angiotensin II blockade reverses myocardial fibrosis in a transgenic mouse model of human hypertrophic cardiomyopathy. Circulation. 2001;103:789-791.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Osio A, Tan L, Chen SN, Lombardi R, Nagueh SF, Shete S, Roberts R, Willerson JT, Marian AJ. Myozenin 2 is a novel gene for human hypertrophic cardiomyopathy. Circ Res. 2007;100:766-768.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Marian AJ, Yu QT, Workman R, Greve G, Roberts R. Angiotensin-converting enzyme polymorphism in hypertrophic cardiomyopathy and sudden cardiac death. Lancet. 1993;342:1085-1086.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Beohar N, Damaraju S, Prather A, Yu QT, Raizner A, Kleiman NS, Roberts R, Marian AJ. Angiotensin-I converting enzyme genotype DD is a risk factor for coronary artery disease. J Investig Med. 1995;43:275-280.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Roberts R, Sigwart U. Current concepts of the pathogenesis and treatment of hypertrophic cardiomyopathy. Circulation. 2005;112:293-296.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Durand JB, Bachinski LL, Bieling LC, Czernuszewicz GZ, Abchee AB, Yu QT, Tapscott T, Hill R, Ifegwu J, Marian AJ. Localization of a gene responsible for familial dilated cardiomyopathy to chromosome 1q32. Circulation. 1995;92:3387-3389.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Li D, Tapscoft T, Gonzalez O, Burch PE, Quiñones MA, Zoghbi WA, Hill R, Bachinski LL, Mann DL, Roberts R. Desmin mutation responsible for idiopathic dilated cardiomyopathy. Circulation. 1999;100:461-464.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Timchenko LT, Timchenko NA, Caskey CT, Roberts R. Novel proteins with binding specificity for DNA CTG repeats and RNA CUG repeats: implications for myotonic dystrophy. Hum Mol Genet. 1996;5:115-121.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Roberts R, Timchenko NA, Miller JW, Reddy S, Caskey CT, Swanson MS, Timchenko LT. Altered phosphorylation and intracellular distribution of a (CUG)n triplet repeat RNA-binding protein in patients with myotonic dystrophy and in myotonin protein kinase knockout mice. Proc Natl Acad Sci USA. 1997;94:13221-13226.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Brugada R, Tapscott T, Czernuszewicz GZ, Marian AJ, Iglesias A, Mont L, Brugada J, Girona J, Domingo A, Bachinski LL. Identification of a genetic locus for familial atrial fibrillation. N Engl J Med. 1997;336:905-911.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Ahmad F, Gonzalez O, Ramagli L, Xu J, Siciliano MJ, Bachinski LL, Roberts R. Identification and characterization of a novel gene (C4orf5) located on human chromosome 4q with specific expression in cardiac and skeletal muscle. Genomics. 2000;70:347-353.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Roberts R. Mechanisms of disease: Genetic mechanisms of atrial fibrillation. Nat Clin Pract Cardiovasc Med. 2006;3:276-282.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Brugada R, Roberts R. Molecular biology and atrial fibrillation. Curr Opin Cardiol. 1999;14:269-273.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Gollob MH, Green MS, Tang AS, Gollob T, Karibe A, Ali Hassan AS, Ahmad F, Lozado R, Shah G, Fananapazir L. Identification of a gene responsible for familial Wolff-Parkinson-White syndrome. N Engl J Med. 2001;344:1823-1831.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Folmes KD, Chan AY, Koonen DP, Pulinilkunnil TC, Baczkó I, Hunter BE, Thorn S, Allard MF, Roberts R, Gollob MH. Distinct early signaling events resulting from the expression of the PRKAG2 R302Q mutant of AMPK contribute to increased myocardial glycogen. Circ Cardiovasc Genet. 2009;2:457-466.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Natarajan A, Yamagishi H, Ahmad F, Li D, Roberts R, Matsuoka R, Hill S, Srivastava D. Human eHAND, but not dHAND, is down-regulated in cardiomyopathies. J Mol Cell Cardiol. 2001;33:1607-1614.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Ahmad F, Li D, Karibe A, Gonzalez O, Tapscott T, Hill R, Weilbaecher D, Blackie P, Furey M, Gardner M. Localization of a gene responsible for arrhythmogenic right ventricular dysplasia to chromosome 3p23. Circulation. 1998;98:2791-2795.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Li D, Ahmad F, Gardner MJ, Weilbaecher D, Hill R, Karibe A, Gonzalez O, Tapscott T, Sharratt GP, Bachinski LL. The locus of a novel gene responsible for arrhythmogenic right-ventricular dysplasia characterized by early onset and high penetrance maps to chromosome 10p12-p14. Am J Hum Genet. 2000;66:148-156.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Li D, Bachinski LL, Roberts R. Genomic organization and isoform-specific tissue expression of human NAPOR (CUGBP2) as a candidate gene for familial arrhythmogenic right ventricular dysplasia. Genomics. 2001;74:396-401.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Mares A Jr, Ledbetter SA, Ledbetter DH, Roberts R, Hejtmancik JF. Isolation of a human chromosome 14-only somatic cell hybrid: analysis using Alu and LINE-based PCR. Genomics. 1991;11:215-218.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Bies RD, Friedman D, Roberts R, Perryman MB, Caskey CT. Expression and localization of dystrophin in human cardiac Purkinje fibers. Circulation. 1992;86:147-153.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Bies RD, Phelps SF, Cortez MD, Roberts R, Caskey CT, Chamberlain JS. Human and murine dystrophin mRNA transcripts are differentially expressed during skeletal muscle, heart, and brain development. Nucleic Acids Res. 1992;20:1725-1731.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Marian AJ, Wu Y, Lim DS, McCluggage M, Youker K, Yu QT, Brugada R, DeMayo F, Quinones M, Roberts R. A transgenic rabbit model for human hypertrophic cardiomyopathy. J Clin Invest. 1999;104:1683-1692.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Patel R, Nagueh SF, Tsybouleva N, Abdellatif M, Lutucuta S, Kopelen HA, Quinones MA, Zoghbi WA, Entman ML, Roberts R. Simvastatin induces regression of cardiac hypertrophy and fibrosis and improves cardiac function in a transgenic rabbit model of human hypertrophic cardiomyopathy. Circulation. 2001;104:317-324.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Tsybouleva N, Zhang L, Chen S, Patel R, Lutucuta S, Nemoto S, DeFreitas G, Entman M, Carabello BA, Roberts R. Aldosterone, through novel signaling proteins, is a fundamental molecular bridge between the genetic defect and the cardiac phenotype of hypertrophic cardiomyopathy. Circulation. 2004;109:1284-1291.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Roberts R. Personalized medicine: a reality within this decade. J Cardiovasc Transl Res. 2008;1:11-16.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  McPherson R, Pertsemlidis A, Kavaslar N, Stewart A, Roberts R, Cox DR, Hinds DA, Pennacchio LA, Tybjaerg-Hansen A, Folsom AR. A common allele on chromosome 9 associated with coronary heart disease. Science. 2007;316:1488-1491.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Preuss M, König IR, Thompson JR, Erdmann J, Absher D, Assimes TL, Blankenberg S, Boerwinkle E, Chen L, Cupples LA. Design of the Coronary ARtery DIsease Genome-Wide Replication And Meta-Analysis (CARDIoGRAM) Study: A Genome-wide association meta-analysis involving more than 22 000 cases and 60 000 controls. Circ Cardiovasc Genet. 2010;3:475-483.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Teslovich TM, Musunuru K, Smith AV, Edmondson AC, Stylianou IM, Koseki M, Pirruccello JP, Ripatti S, Chasman DI, Willer CJ. Biological, clinical and population relevance of 95 loci for blood lipids. Nature. 2010;466:707-713.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Schunkert H, König IR, Kathiresan S, Reilly MP, Assimes TL, Holm H, Preuss M, Stewart AF, Barbalic M, Gieger C. Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nat Genet. 2011;43:333-338.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Jarinova O, Stewart AF, Roberts R, Wells G, Lau P, Naing T, Buerki C, McLean BW, Cook RC, Parker JS. Functional analysis of the chromosome 9p21.3 coronary artery disease risk locus. Arterioscler Thromb Vasc Biol. 2009;29:1671-1677.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Soranzo N, Spector TD, Mangino M, Kühnel B, Rendon A, Teumer A, Willenborg C, Wright B, Chen L, Li M. A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium. Nat Genet. 2009;41:1182-1190.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Dandona S, Chen L, Fan M, Alam MA, Assogba O, Belanger M, Williams K, Wells GA, Tang WH, Ellis SG. The transcription factor GATA-2 does not associate with angiographic coronary artery disease in the Ottawa Heart Genomics and Cleveland Clinic GeneBank Studies. Hum Genet. 2010;127:101-105.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Dandona S, Stewart AF, Roberts R. Genomics in coronary artery disease: past, present and future. Can J Cardiol. 2010;26 Suppl A:56A-59A.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Dandona S, Stewart AF, Chen L, Williams K, So D, O'Brien E, Glover C, Lemay M, Assogba O, Vo L. Gene dosage of the common variant 9p21 predicts severity of coronary artery disease. J Am Coll Cardiol. 2010;56:479-486.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Pare G. Genome-wide association studies--data generation, storage, interpretation, and bioinformatics. J Cardiovasc Transl Res. 2010;3:183-188.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Reilly MP, Li M, He J, Ferguson JF, Stylianou IM, Mehta NN, Burnett MS, Devaney JM, Knouff CW, Thompson JR. Identification of ADAMTS7 as a novel locus for coronary atherosclerosis and association of ABO with myocardial infarction in the presence of coronary atherosclerosis: two genome-wide association studies. Lancet. 2011;377:383-392.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Rafiq S, Anand S, Roberts R. Genome-wide association studies of hypertension: have they been fruitful? J Cardiovasc Transl Res. 2010;3:189-196.  [PubMed]  [DOI]  [Cited in This Article: ]