ISSN: 1885-5857 Impact factor 2023 7.2
Vol. 62. Num. 3.
Pages 305-313 (March 2009)

Genomics and Proteomics in Heart Failure Research

La genómica y la proteómica en la investigación de la insuficiencia cardiaca

Arantxa GonzálezaBegoña LópezaJavier BeaumontaSusana RavassaaTeresa AriasaNerea HermidaaAmaia ZudaireaJavier Díezb

Options

Heart failure is a complex syndrome and is one of the main causes of morbidity and mortality in developed countries. Despite considerable research effort in recent years, heart failure prevention and treatment strategies still suffer significant limitations. New theoretical and technical approaches are, therefore, required. It is in this context that the «omic» sciences have a role to play in heart failure. The incorporation of «omic» methodologies into the study of human disease has substantially changed biological approaches to disease and has given an enormous impetus to the search for new disease mechanisms, as well as for novel biomarkers and therapeutic targets. The application of genomics, proteomics and metabonomics to heart failure research could increase our understanding of the origin and development of the different processes contributing to this syndrome, thereby enabling the establishment of specific diagnostic profiles and therapeutic templates that could help improve the poor prognosis associated with heart failure. This brief review contains a short description of the fundamental principles of the «omic» sciences and an evaluation of how these new techniques are currently contributing to research into human heart failure. The focus is mainly on the analysis of gene expression microarrays in the field of genomics and on studies using two-dimensional electrophoresis with mass spectrometry in the area of proteomics.

Keywords

Genomics
Proteomics
Heart failure

INTRODUCTION

Heart failure, a complex syndrome the natural history of which leads to left ventricular dysfunction (both systolic and diastolic),1 is one of the main causes of morbidity and mortality in the Western world, and particularly so in Spain.2 Owing to the aging of the population and the greater longevity of patients with cardiovascular disease, the incidence and prevalence of heart failure is increasing.3 Should this trend continue, the next 20 years will see heart failure reach epidemic proportions; such a bleak future has been predicted for Spain.4 Given the high medical and health system costs associated with this problem, heart failure has become the cardiovascular disease of greatest economic burden in this country.5

Despite the enormous efforts of the last 50 years to understand the pathophysiology and pharmacology of heart failure, a number of recently published studies highlight the important limitations suffered with respect to its prevention and treatment— factors that explain the poor prognosis associated with this condition.6 New conceptual and practical approaches are therefore needed,7 among which the "omic" focus on heart failure finds its place.

"Omics" refers to the biological sciences that study the genes, their initial products (RNA transcripts), their final products (proteins) and the metabolites produced in the processes in which these proteins are involved. The incorporation of omic methodology into the study of human disease has substantially modified the biological focus placed upon them (Figure 1), and has greatly stimulated research into mechanisms, biomarkers, and therapeutic targets. Although the molecular causes of heart dysfunction that lead to heart failure are still incompletely understood, it is thought that they involve abnormal gene expression and protein production. It has been proposed that the combination of the different "omic" sciences, especially genomics and proteomics, might help in our understanding of the origin and development of the different entities that make up the syndrome of heart failure, and therefore in the establishment of diagnostic profiles and therapeutic patterns that might improve the poor prognosis with which this problem is associated.8

Figure 1. A new view of human disease from the point of view of the omic sciences. 1: transcription; 2: translation; 3: post-translational modifications; 4: interactions; 5: integration.

BASIC CONCEPTS OF GENOMICS AND PROTEOMICS

Genomics

Genomics is the study of the molecular organization of DNA and its physical mapping. Genomics encompasses different subspecialties. For example, structural genomics studies the folding of macromolecules and their three dimensional structure with the aid of techniques derived from physics (eg, x-ray crystallography) and bioinformatics, and classifies these molecules into functional families. Biochemical genomics studies groups of specific proteins and their corresponding open reading frames (ORF). A large number of proteins has been purified, mapped to a specific ORF, and classified.9 Chemical genomics studies small molecules to determine their possible modulatory effects on cell status or gene expression; this is preferably undertaken making use of high performance systems.10 Functional or physiological genomics focuses on the functional analysis of the entire genome, the identification of the structure of DNA and the characterization of its molecular function, as well as the interactions between genes and their products.11,12 Epigenomics examines the interactions between the genome and the proteome along with the general patterns of methylation and methylation signals, and compares this information among species. Comparative genomics or phylogenomics tries to determine the number of protein families encoded by different genomes, the distribution of coding genes in different genomes, and how many genes are shared by different genomes.13 Orthogenomics studies the genomes of orthologous descendants, while paragenomics studies paralogous genomes. Both latter types of study try to determine the composition and organization of protein domains in different organisms. Genetic genomics includes the analysis of expression profiles and of fingerprints based on markers in each individual belonging to a segregating population. Computational genomics measures (qualitatively and quantitatively) properties of interest, for example in different strains of mouse. Gene variation can then be identified and correlated to phenotypic traits in the different strains. The degree of correlation between these features and the grouping of the strains within each haplotype block is examined by ANOVA.14 The aim of nutrigenomics is to determine the effects of macro- and micronutrients in health and disease in different genotypes.15 Toxicogenomics aims to understand the complexities of biological systems in their responses to toxins, mutagens, and carcinogens. Transcriptomics deserves special mention because of its great functional importance. This subspecialty focuses on the study of the transcriptome of cells or organs in determined situations, including disease. The transcriptome includes the collection of RNA molecules transcribed from the genome, ie, the expression profile of messenger RNA (mRNA). In any human cell, about one half of all the genes might be expressing at any given time, and at the organism level (again in humans), some 25 000-30 000 genes are expressed. It has been calculated that the total number of transcripts from these genes in different types of cell is 134 135. While the mean copy number of transcripts for some cell types is just 0.3, in others it is 9417.

After completing the sequencing of the genome of an organism, interest centers on the functions of the genes. The studies such interest demands can now be performed thanks to the development of high performance procedures16 that can detect thousands of ORF. This area of study integrates the disciplines of genetics, molecular biology, biochemistry, pharmacology (which gives rise to pharmacogenomics or the design of drugs better adjusted to the genetic constitution of the individual), agriculture, medicine, and others.

Proteomics

Proteomics is defined as the study of the proteome, ie, the entire set of proteins present in a cell at any given moment in time. The genome does not reveal the details of functioning within a cell; this is the domain of the proteomics. The proteome shows variations depending on the stage of development, the organ in question, the metabolic rate, and the health of the organism etc.

Since proteins are expressed and organized within systems that interact, their study can be very complicated. Large scale proteomic studies may involve examining the composition and structure of proteins, identifying their isoforms, examining conformational changes and modulatory changes during development, studying post-transcriptional and post-translational changes (phosphorylation, glycosylation, etc), and examining the interactions between proteins or proteins and drugs etc. Large numbers of proteins can be identified even in very small samples. Proteomics has changed the focus of research into biological functions. Unlike traditional experimental designs based on the need of a starting hypothesis, proteomic technology allows more direct approximations to be made based on the study of networks of molecular interactions.

Proteomics also has a number of subspecialties. Expression proteomics analyzes cellular proteins by 2-dimensional electrophoresis or high resolution chromatography combined with mass spectrometry.17 Proteomic cartography deals with the interaction between proteins in different phases of cell function.18 Functional proteomics focuses on the study of specific protein functions.19 Structural proteomics tries to understand cell functioning based on 3-dimensional analysis and the construction of protein models.20,21 Inverse proteomics studies proteins via their corresponding genes.

Related to proteomics is metabolomics, which studies the metabolic status of fluids and animal tissue preparations using high resolution nuclear magnetic resonance spectroscopy and statistics. The study of the entire set of metabolites present at any moment can provide important information on the phenotype of an organism. Metabolites are the true final products of gene transcription and reflect in the most exact way the activity of a cell or its functional phenotype. Therefore, it is probable that the metabolome affords the most adequate scenario for the study of cellular processes in both physiological and pathological situations.22 Metabolomic studies can be of use in the development of personalized drug treatments if the differences between the metabolic phenotype before and after the administration of the drug can be established. Such information would help predict individual responses.23

THE GENOMICS OF HEART FAILURE

Heart failure is ideally suited to genomic study since it is a complex syndrome with multiple etiologies and predisposing factors (both environmental and genetic). While the traditional approach based on the study of a small number of genes or a single risk factor can generate partial information regarding this disease, the genomic approach, which identifies large numbers of genes and abnormal signaling pathways in their different stages of development, is much more efficient, accelerating our understanding of the processes involved.24

Gene expression microarrays provide qualitative (identification of active and silenced genes) and quantitative (the expression level) information on gene functioning, allowing subtle changes in gene functioning to be identified. These microarrays have been of great use in oncology, helping in the diagnosis and classification of tumors,25 the

identification of abnormal signaling pathways, in making contributions to our pathophysiological knowledge of cancer, and helping in the development of more specialized therapies.26 Similarly, using microarrays in heart failure research could help differentiate between different molecular subtypes of disease.

Commercially available and specially designed microarrays (for specific heart genes) have both been used in human studies. To date, expression studies have been performed involving small populations in order to compare patients with heart failure to persons without this problem, and on samples of myocardium before and after the implantation of a device to assist the left ventricle. The patients involved in these studies therefore represent advanced cases of heart failure. Table shows the main findings from a number of these studies.27-44 The expression of hundreds of genes involved in calcium-mediated cell signaling, energy metabolism, apoptosis, signal transduction, transmembrane ion transport, and the maintenance of the cytoskeleton and the extracellular matrix was found to be altered.

A possible clinical application of this type of study is the classification of different cardiomyopathies and the prediction of responses to treatment. For example, a few studies have tried to distinguish the etiology of heart failure and dilated cardiomyopathy (DCM) by defining their characteristic expression profiles.34,36,37,44 In this context, the analysis of gene expression allowed the distinction to be made between patients with cardiomyopathy linked to alcohol consumption and those with familial cardiomyopathy,32 and between patients with DCM and those with ischemic cardiomyopathy.39 Such studies have also shown great differences to exist in the gene expression profiles of patients with hypertrophic cardiomyopathy and those with DCM in the advanced stages heart failure.43 The use of microarrays has also made it possible to distinguish between an ischemic and non-ischemic origin of heart failure with 89% sensitivity and specificity.45 However, in a recent study it was impossible to clearly distinguish between cardiomyopathy of ischemic and non-ischemic origin.46 Thus, although the results seem promising, the true clinical usefulness of gene expression microarrays for the classification and prognosis of different cardiomyopathies requires validation in large populations of patients (the majority of studies performed so far have involved only small numbers).

Work is also underway investigating the use of expression profiles as prognostic transcriptomic biomarkers. In a recent study involving 108 patients with newly diagnosed DCM, and in which patients with good and poor prognoses were compared after more than five years of follow-up, a panel of 45 genes were identified that were overexpressed in the patients with a good prognosis. This panel of genes was invested with a sensitivity of 74% and a specificity of 90% with regard to predictions of patient evolution.47

Although the application of pharmacogenomics to heart failure is in its infancy, clinical research has shown that certain neurohumoral polymorphisms modify the effectiveness of different drugs and the course of disease in patients. Such is the case of functional polymorphisms of the genes of the reninangiotensin system, the sympathetic nervous system, and the gene for endothelial nitric oxide synthetase, all of which affect the concentration of effectors of these systems and their signaling pathways.48

The assessment of cardiac genomic information is not without its limitations. In particular, the factors that can influence the variability of the results obtained are many. The area of the organ from where a tissue is taken may influence the results,30,49 variables such as age and sex may certainly affect the gene expression profile,34,50 and the impact of pharmacological treatment and concomitant disease, etc, also needs to be considered. Thus, although microarray technology and its capacity for analyzing thousands of genes at the same time is providing interesting results in some areas, more solid evidence is required to back up the use of the technique in general clinical practice.

THE PROTEOMICS OF HEART FAILURE

Currently, four databases for human cardiac proteins exist, all based on information from 2-dimensional gel studies (Figure 2). These databases were established by three independent groups of researchers and are available on the Web. All meet the standards established for databases based on information from 2-dimensional gels.51 Databases are also being established for other mammals,52,53 thus accelerating work in animal models of heart failure.

Figure 2. Image of a 2-dimensional gel running a sample of human whole left ventricular tissue (visualized with SYPRO Ruby), showing the species of proteins localizing according to their molecular weight and pH.

The analysis of the changes that occur in the proteome of the cells of the left ventricle during progress towards heart failure have revealed great differences to exist between expression patterns in different heart problems.54-56 However, the majority of proteins whose expression is altered are involved with inflammation, calcium-mediated signaling, cell growth and death pathways, the cytoskeleton, and the extracellular matrix. Most studies have focused on DCM, and a few on ischemic heart disease , and to date the expression of more than 100 proteins has been found altered (as determined in 2-dimensional gel analyses) in patients with these disease. The majority are downregulated in patients with heart failure.57-62 The use of mass spectrometry has shown that many of these proteins belong to the cytoskeleton and the myofilaments, to be associated with the mitochondria and energy production, and to be related to stress responses.63 For example, in the context of stress proteins, 59 isoelectric isoforms of heat shock protein 27 (HSP27) have been found in human myocardium. The expression of 12 of these is quantitatively altered in the hearts of patients with DCM, as are 10 in the myocardium of patients with heart failure of ischemic origin.61

With respect to studies in plasma, the work of the Human Proteomic Organization (HUPO) (whose aim it is to systematically analyze the subproteome of human plasma) stands out. Initial studies in heart failure detected alterations in the expression of families of proteins involved in inflammation, growth, differentiation, intracellular signaling, the cytoskeleton, channels and receptors, and myocardial remodelling.64 The analysis of the plasma subproteome could also be of prognostic use. For example, the analysis of circulating proteins in the plasma using SELDI-TOF protein chip technology has allowed the identification of proteins associated with the remodeling of the left ventricle following an acute myocardial infarction. This could help identify patients at greater risk of heart failure associated with such an event.65

In the area of metabolomics, studies related to heart failure have centered on the search for biomarkers of disease progress. In a recent study, the serum metabolome of a group of patients with heart failure and a reduced left ventricular ejection fraction was studied. Among the many metabolites altered, 2-oxoglutarate and pseudouridine were examined in detail. Studying this combination provided a more sensitive and more specific diagnosis of heart failure than monitoring brain natriuretic peptide levels.66

PERSPECTIVES

The conviction exists among researchers that simply knowing the complete sequence of the genome, the transcriptional changes and expression levels of thousands of genes, and the post-translational changes and functional variations of thousands of proteins, will simply not be enough to elucidate the etiopathogenesis and pathophysiology of disease. The consensus is that the true challenge lies in the appropriate handling of such vast quantities of information. Generating instruments that can efficiently analyze this data and assess it effectively is therefore just as important as generating the information itself. High performance bioinformatics and computational biology, respectively, are the instruments that try to meet these needs. Both are based on systems biology, ie, the construction of networks of genes, proteins and metabolic pathways that interact to form functional modules (Figure 3). In turn, these modules are integrated into models designed to predict clinical phenotypes, diagnostic strategies, and therapeutic strategies after completing the appropriate experimental studies.

Figure 3. Steps followed in systems biology from the generation of molecular and biochemical data through to assessment and basic and clinical experimental validation.

Systems biology of the heart is still in its infancy.67 To date, models have been developed that integrate genomic and proteomic information and the function of organelles such as the mitochindrion68 or entire cardiomyocetes.69 With respect to heart failure, the first attempt at systematization has already been made, with the development of clinical diagnostic models from the changes detected in the expression of genes in patients with DCM and other heart problems.70 In the near future more holistic models may become available that integrate information regarding the cardiac genome through to the behavior of heart diseases, making the personalized management of patients with heart failure possible.

Section Sponsored by Labotatorio Dr Esteve


Correspondence: Dr. J. Díez.
Área de Ciencias Cardiovasculares. Edificio CIMA. Avda. Pío XII, 55. 31008 Pamplona. Navarra. España.
E-mail: jadimar@unav.es

Bibliography
[1]
Heart Failure Society of America (HFSA) practice guidelines..
HFSA guidelines for management of patients with heart failure caused by left ventricular systolic dysfunction-pharmacological approaches..
J Card Fail, (1999), 5 pp. 354-82
[2]
Rodríguez-Artalejo F, Guallar-Castellón P, Banegas-Banegas JR, del Rey Calero..
Trends in hospitalization and mortality for heart failure in Spain..
Eur Heart J, (1997), 18 pp. 1771-9
[3]
Anguita Sánchez M, Crespo Leiro M, de Teressa Galván E, Jiménez Navarro M, Alonso-Pulpón L, Muñiz García J, en nombre de los investigadores del estudio PRICE..
Prevalencia de la insuficiencia cardiaca en la población general española mayor de 45 años. Estudio PRICE..
Rev Esp Cardiol, (2008), 61 pp. 1041-9
[4]
Sáez T, Suárez C, Blanco F, Gabriel R..
Epidemiología de las enfermedades cardiovasculares en la población anciana española..
Rev Esp Cardiol, (1998), 51 pp. 864-73
[5]
Sala J, Marrugat J, Roure J, Cosín-Aguilar J..
The management of heart failure in Spain..
Eur J Heart Fail, (2000), 2 pp. 341-4
[6]
MacIntyre K, Capewell S, Stewart S, Chalmers JW, Boyd J, Finlayson A, et al..
Evidence of improving prognosis in heart failure. Trends in case fatality in 66547 patients hospitalized between 1986 and 1995..
Circulation, (2000), 102 pp. 1126-31
[7]
Katz AM..
The modern view of heart failure..
Circulation Heart Fail, (2008), 1 pp. 63-71
[8]
Dos Remedios CG, Liew CC, Allen PD, Winslow RL, van Eyk JE, Dunn MJ..
Genomics, proteomics and bioinformatics of human heart failure..
J Muscle Res Cell Motil, (2003), 24 pp. 251-60
[9]
Martzen MR, McCraith SM, Spinelli SL, Torres FM, Fields S, Grayhack EJ, et al..
A biochemical genomics approach for identifying genes by the activity of their products..
Science, (1999), 286 pp. 1153-5
[10]
Stegmaier K, Ross KN, Colavito SA, O'Malley S, Stockwell BR, Golub TR..
Gene expression-based high-throughput screening (GE-HTS) and application to leukemia differentiation..
Nature Genet, (2004), 36 pp. 257-63
[11]
Wu LF, Hughes TR, Davierwala AP, Robinson MD, Stoughton R, Altschuler SJ..
Large-scale prediction of Saccharomyces cerevisiae gene function using overlapping transcriptional clusters..
Nature Genet, (2002), 31 pp. 255-65
[12]
Liang P, Labedan B, Riley M..
Physiological genomics of Escherichia coli protein families..
Physiol Genomics, (2002), 9 pp. 15-26
[13]
Ureta-Vidal A, Ettwiller L, Birney E..
Comparative genomics: genome-wide analysis in metazoan eukaryotes..
Nature Rev Genet, (2003), 4 pp. 251-62
[14]
Wang J, Liao G, Usuka J, Peltz G..
Computational genetics:from mouse to human? Trends Genet, (2005), 21 pp. 526-32
[15]
Müller M, Kersten S..
Nutrigenomics: goals and strategies..
Nature Rev Genet, (2003), 4 pp. 315-22
[16]
Ross-Macdonald P, Coelho PS, Roemer T, Agarwal S, Kumar A, Jansen R, et al..
Large-scale analysis of the yeast genome by transposon tagging and gene disruption..
Nature, (1999), 402 pp. 413-8
[17]
Wagner K, Miliotis T, Marko-Varga G, Bischoff R, Unger KK..
An automated on-line multidimensional HPLC system for protein and peptide mapping with integrated sample preparation..
Anal Chem, (2002), 74 pp. 809-20
[18]
Blackstock WP, Weir MP..
Proteomics: quantitative and physical mapping of cellular proteins..
Trends Biotechnol, (1999), 17 pp. 121-7
[19]
Graves PR, Haystead TA..
Molecular biologist's guide to proteomics..
Microbiol Mol Biol Rev, (2002), 66 pp. 39-63
[20]
Norin M, Sundstrom M..
Structural proteomics: developments in structure-to-function predictions..
Trends Biotechnol, (2002), 20 pp. 79-84
[21]
Sali A, Glaeser R, Earnest T, Baumeister W..
From words to literature in structural proteomics..
Nature, (2003), 422 pp. 216-25
[22]
van der Greef J, Stroobant P, van der Heijden R..
The role of analytical sciences in medical systems biology..
Curr Opin Chem Biol, (2004), 8 pp. 559-65
[23]
Clayton TA, Lindon JC, Cloarec O, Antti H, Charuel C, Hanton G, et al..
Pharmaco-metabonomic phenotyping and personalized drug treatment..
Nature, (2006), 440 pp. 1073-7
[24]
Liew CC, Dzau VJ..
Molecular genetics and genomics of heart failure..
Nature Rev Genet, (2004), 5 pp. 811-25
[25]
Golub TR, Slonim DK, Tamayo P, Huard C, Gaasenbeek M, Mesirov JP, et al..
Molecular classification of cancer: class discovery and class prediction by gene expression monitoring..
Science, (1999), 286 pp. 531-7
[26]
Bild AH, Yao G, Chang JT, Wang Q, Potti A, Chasse D, et al..
Oncogenic pathway signatures in human cancers as a guide to targeted therapies..
Nature, (2006), 439 pp. 353-7
[27]
Donahue MP, Marchuk DA, Rockman HA..
Redefining heart failure. The utility of genomics..
J Am Coll Cardiol, (2006), 48 pp. 1289-98
[28]
Yang J, Moravec CS, Sussman MA, DiPaola NR, Fu D, Hawthorn L, et al..
Decreased SLIM1 expression and increased gelsolin expression in failing human hearts measured by highdensity oligonucleotide arrays..
Circulation, (2000), 102 pp. 3046-52
[29]
PGC1alpha and ERRalpha target gene downregulation is a signature of the failing human heart. J Mol Cell Cardiol. 2008 [Epub ahead of print].
[30]
Steenman M, Chen YW, Le Cunff M, Lamirault G, Varró A, Hoffman E, et al..
Transcriptomal analysis of failing and nonfailing human hearts..
Physiol Genomics, (2003), 12 pp. 97-112
[31]
Barrans JD, Allen PD, Stamatiou D, Dzau VJ, Liew CC..
Global gene expression profiling of end-stage dilated cardiomyopathy using a human cardiovascular-based cDNA microarray..
Am J Pathol, (2002), 160 pp. 2035-43
[32]
Tan FL, Moravec CS, Li J, Apperson-Hansen C, McCarthy PM, Young JB, et al..
The gene expression fingerprint of human heart failure..
Proc Natl Acad Sci U S A, (2002), 99 pp. 11387-92
[33]
Gene expression profiles in end-stage human idiopathic dilated cardiomyopathy: altered expression of apoptotic and cytoskeletal genes. Genomics. 2004,83:281-97.
[34]
Haddad GE, Saunders LJ, Crosby SD, Carles M, del Monte F, King K, et al..
Human cardiac-specific cDNA array for idiopathic dilated cardiomyopathy: sex-related differences..
Physiol Genomics, (2008), 33 pp. 267-77
[35]
Grzeskowiak R, Witt H, Drungowski M, Thermann R, Hennig S, Perrot A, et al..
Expression profiling of human idiopathic dilated cardiomyopathy..
Cardiovasc Res, (2003), 59 pp. 400-11
[36]
Beisvag V, Lehre PK, Midelfart H, Aass H, Geiran O, Sandvik AK, et al..
Aetiology-specific patterns in end-stage heart failure patients identified by functional annotation and classification of microarray data..
Eur J Heart Fail, (2006), 8 pp. 381-9
[37]
Barth AS, Kuner R, Buness A, Ruschhaupt M, Merk S, Zwermann L, et al..
Identification of a common gene expression signature in dilated cardiomyopathy across independent microarray studies..
J Am Coll Cardiol, (2006), 48 pp. 1610-7
[38]
Steenbergen C, Afshari CA, Petranka JG, Collins J, Martin K, Bennett L, et al..
Alterations in apoptotic iguresin in human idiopathic cardiomyopathic hearts in failure..
Am J Physiol Heart Circ Physiol, (2003), 284 pp. H268-76
[39]
Blaxall BC, Tschannen-Moran BM, Milano CA, Koch WJ..
Differential gene expression and genomic patient stratification following left ventricular assist device support..
J Am Coll Cardiol, (2003), 41 pp. 1096-106
[40]
Chen Y, Park S, Li Y, Missov E, Hou M, Han X, et al..
Alterations of gene expression in failing myocardium following left ventricular assist device support..
Physiol Genomics, (2003), 14 pp. 251-60
[41]
Hall JL, Grindle S, Han X, Fermin D, Park S, Chen Y, et al..
Genomic profiling of the human heart before and after mechanical support with a ventricular assist device reveals alterations in vascular iguresin networks..
Physiol Genomics, (2004), 17 pp. 283-91
[42]
Hall JL, Birks EJ, Grindle S, Cullen ME, Barton PJ, Rider JE, et al..
Molecular signature of recovery following combination left ventricular assist device (LVAD) support and pharmacologic therapy..
Eur Heart J, (2007), 28 pp. 613-27
[43]
Hwang JJ, Allen PD, Tseng GC, Lam CW, Fananapazir L, Dzau VJ, et al..
Microarray gene expression profiles in dilated and hypertrophic cardiomyopathic end-stage heart failure..
Physiol Genomics, (2002), 10 pp. 31-44
[44]
Ruppert V, Meyer T, Pankuweit S, Möller E, Funck RC, Grimm W, et al..
Gene expression profiling from endomyocardial biopsy tissue allows distinction between subentities of dilated cardiomyopathy..
J Thorac Cardiovasc Surg, (2008), 136 pp. 360-9
[45]
Kittleson MM, Ye SQ, Irizarry RA, Minhas KM, Edness G, Conte JV, et al..
Identification of a gene expression profile that differentiates between ischemic and nonischemic cardiomyopathy..
Circulation, (2004), 110 pp. 3444-51
[46]
Kuner R, Barth AS, Ruschhaupt M, Buness A, Zwermann L, Kreuzer E, et al..
Genomic analysis reveals poor separation of human cardiomyopathies of ischemic and nonischemic etiologies..
Physiol Genomics, (2008), 34 pp. 88-94
[47]
Heidecker B, Kasper EK, Wittstein IS, Champion HC, Breton E, Russell SD, et al..
Transcriptomic biomarkers for individual risk assessment in new-onset heart failure..
Circulation, (2008), 118 pp. 238-46
[48]
McNamara DM..
Pharmacogenomics for neurohormonal intervention in heart failure..
Cardiol Clin, (2008), 26 pp. 127-35
[49]
Thum T, Borlak J..
Gene expression in distinct regions of the heart..
[50]
Boheler KR, Volkova M, Morrell C, Garg R, Zhu Y, Margulies K, et al..
Sex-and age-dependent human transcriptome variability: implications for chronic heart failure..
Proc Natl Acad Sci U S A, (2003), 100 pp. 2754-9
[51]
Federated two-dimensional electrophoresis database: a simple means of publishing two-dimensional electrophoresis data. Electrophoresis.1996;17:540-6.
[52]
Li XP, Pleissner KP, Scheler C, Regitz-Zagrosek V, Salnikow J, Jungblut PR..
A two-dimensional electrophoresis database of rat heart proteins..
[53]
Dunn MJ, Corbett JM, Wheeler CH..
HSC-2DPAGE and the two-dimensional gel electrophoresis database of dog heart proteins..
Electrophoresis, (1997), 18 pp. 2795-802
[54]
Gramolini AO, Kislinger T, Liu PP, MacLenna DH, Emily A..
Analyzing the cardiac muscle proteome by liquid chromatography-mass spectrometry (LC-MS) based expression proteomics..
Meth Mol Biol, (2006), 357 pp. 15-32
[55]
McGregor E, Dunn MJ..
Proteomics of the heart. Unraveling disease..
[56]
Arab S, Gramolini AO, Ping P, Kisslinger T, Stanley B, Van Eyk J, et al..
Cardiovascular proteomics. Tools to develop new biomarkers and potential applications..
J Am Coll Cardiol, (2006), 48 pp. 1733-41
[57]
Corbett JM, Why HJ, Wheeler CH, Richardson PJ, Archard LC, Yacoub MH, et al..
Cardiac protein abnormalities in dilated cardiomyopathy detected by two-dimensional polyacrylamide gel electrophoresis..
Electrophoresis, (1998), 19 pp. 2031-42
[58]
Knecht M, Regitz-Zagrosek V, Pleissner KP, Emig S, Jungblut P, Hildebrandt A, et al..
Dilated cardiomyopathy: computerassisted analysis of endomyocardial biopsy protein patterns by two-dimensional gel electrophoresis..
Eur J Clin Chem Clin Biochem, (1994), 32 pp. 615-24
[59]
Knecht M, Regitz-Zagrosek V, Pleissner KP, Jungblut P, Steffen C, Hildebrandt A, et al..
Characterization of myocardial protein composition in dilated cardiomyopathy by twodimensional gel electrophoresis..
Eur Heart J, (1994), 15 pp. 37-44
[60]
Pleissner KP, Regitz-Zagrosek V, Weise C, Neuss M, Krüdewagen B, Söding P, et al..
Chamber-specific expression of human myocardial proteins detected by two-dimensional gel electrophoresis..
Electrophoresis, (1995), 16 pp. 841-50
[61]
Scheler C, Li XP, Salnikow J, Dunn MJ, Jungblut PR..
Comparison of two-dimensional electrophoresis patterns of heat shock protein Hsp27 species in normal and cardiomyopathic hearts..
[62]
Banfi C, Brioschi M, Wait R, Begum S, Gianazza E, Fratto P, et al..
Proteomic analysis of membrane microdomains derived from both failing and non-failing human hearts..
Proteomics, (2006), 6 pp. 1976-88
[63]
McGregor E, Dunn MJ..
Proteomics of heart disease..
Hum Mol Genet, (2003), 12 pp. R135-44
[64]
Berhane BT, Zong C, Liem DA, Huang A, Le S, Edmondson RD, et al..
Cardiovascular-related proteins identified in human plasma by the HUPO Plasma Proteome Project pilot phase..
Proteomics, (2005), 5 pp. 3520-30
[65]
Pinet F, Beseme O, Cieniewski-Bernard C, Drobecq H, Jourdain S, Lamblin N, et al..
Predicting left ventricular remodelling after a first myocardial infarction by plasma proteome analysis..
Proteomics, (2008), 8 pp. 1798-808
[66]
Dunn WB, Broadhurst DI, Deepak SM, Buch MH, McDowell G, Spasic I, et al..
Serum metabolomics reveals many novel metabolic markers of heart failure, including pseudouridine and 2-oxoglutarate..
Metabolomics, (2007), 3 pp. 413-26
[67]
Shreenivasaiah PK, Rho S-H, Kim T, Kim DH..
An overview of cardiac systems biology..
J Mol Cell Cardiol, (2008), 44 pp. 460-9
[68]
Vo TD, Palsson BO..
Building the power house: recent advances in mitochondrial studies therough proteomics and systems biology..
Am J Physiol Cell Physiol, (2007), 292 pp. C164-77
[69]
Cortassa S, Aon M, O'Rourke B, Jacques R, Tseng HJ, Marban E..
A computacional model integrating electrophysiology, contraction, and mitochondrial bioenergetics in the ventricular myocyte..
Biophys J, (2006), 91 pp. 1564-89
[70]
Camargo A, Azuaje F..
Identification of dilated cardiomyopathy signature genes through gene expression and network data integration..
Genomics, (2008), 92 pp. 404-13
Are you a healthcare professional authorized to prescribe or dispense medications?