Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Diabetes ; 70(1): 255-261, 2021 01.
Article in English | MEDLINE | ID: mdl-33115826

ABSTRACT

The incidence of atrial fibrillation (AF) is higher in patients with diabetes. The goal of this study was to assess if the addition of plasma lipids to traditional risk factors could improve the ability to detect and predict future AF in patients with type 2 diabetes. Logistic regression models were used to identify lipids associated with AF or future AF from plasma lipids (n = 316) measured from participants in the ADVANCE trial (n = 3,772). To gain mechanistic insight, follow-up lipid analysis was undertaken in a mouse model that has an insulin-resistant heart and is susceptible to AF. Sphingolipids, cholesteryl esters, and phospholipids were associated with AF prevalence, whereas two monosialodihexosylganglioside (GM3) ganglioside species were associated with future AF. For AF detection and prediction, addition of six and three lipids, respectively, to a base model (n = 12 conventional risk factors) increased the C-statistics (detection: from 0.661 to 0.725; prediction: from 0.674 to 0.715) and categorical net reclassification indices. The GM3(d18:1/24:1) level was lower in patients in whom AF developed, improved the C-statistic for the prediction of future AF, and was lower in the plasma of the mouse model susceptible to AF. This study demonstrates that plasma lipids have the potential to improve the detection and prediction of AF in patients with diabetes.


Subject(s)
Atrial Fibrillation/diagnosis , Diabetes Mellitus, Type 2/complications , Lipids/blood , Aged , Animals , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Male , Mice , Middle Aged , Risk Assessment , Risk Factors
2.
Adv Exp Med Biol ; 1229: 343-354, 2020.
Article in English | MEDLINE | ID: mdl-32285423

ABSTRACT

Heart failure is the end result of a variety of cardiovascular disease states. Heart failure remains a challenge to treat, and the incidence continues to rise with an aging population, and increasing rates of diabetes and obesity. Non-coding RNAs, once considered as "junk DNA", have emerged as powerful transcriptional regulators and potential therapeutic targets for the treatment of heart failure. Different classes of non-coding RNAs exist, including small non-coding RNAs, referred to as microRNAs, and long non-coding RNAs. Both microRNAs and long non-coding RNAs play a role in cardiac development as well as in the pathogenesis of cardiovascular disease, prompting many studies to investigate their role as potential therapeutic targets. Most studies manipulate miRNAs and lncRNAs of interest via antisense oligonucleotides; however, several challenges remain limiting their potential clinical value. As such, viral and non-viral delivery methods are being developed to achieve targeted delivery in vivo.


Subject(s)
Cardiovascular Diseases/therapy , RNA, Untranslated , Translational Research, Biomedical , Aging , Cardiovascular Diseases/genetics , Heart Failure/genetics , Heart Failure/therapy , Humans , MicroRNAs , RNA, Long Noncoding
3.
Arch Toxicol ; 94(5): 1763-1768, 2020 05.
Article in English | MEDLINE | ID: mdl-32172307

ABSTRACT

Open-access gene expression data sets provide a useful resource for identifying novel drug targets and biomarkers. The IGF1-PI3K pathway is a critical mediator of physiological cardiac enlargement/hypertrophy and protection. This study arose after mining a gene microarray data set from a previous study that compared heart tissue from cardiac-specific PI3K transgenic mouse models. The top-ranked candidate identified from the microarray data was clusterin. Clusterin has been proposed as a biomarker for multiple diseases including heart failure, and as a cancer drug target. Here, we show that clusterin gene expression is increased in hearts of transgenic mice with increased PI3K and decreased in mice with depressed cardiac PI3K. In vitro, clusterin secretion was elevated in media from neonatal rat ventricular myocytes treated with IGF1. Furthermore, by mining gene expression data from hearts during normal mouse postnatal growth, we also report an increase in clusterin expression with postnatal heart growth. Given we show that clusterin is regulated by the IGF1-PI3K pathway in the heart, and this pathway mediates physiological cardiac hypertrophy and cardioprotection, caution is required when considering clusterin as a biomarker for heart failure and as a cancer target. Mining pre-existing cardiac profiling data sets may be a useful approach to assess whether regulating new drug targets is likely to lead to cardiac damage/toxicity.


Subject(s)
Cardiotoxicity , Clusterin/metabolism , Drug Delivery Systems , Myocardium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Cardiomegaly , Heart Failure , Insulin-Like Growth Factor I , Mice , Mice, Transgenic , Myocytes, Cardiac , Pharmaceutical Preparations , Rats , Signal Transduction
4.
Cell Rep ; 24(10): 2757-2772, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30184508

ABSTRACT

Exercise-induced heart growth provides protection against cardiovascular disease, whereas disease-induced heart growth leads to heart failure. These distinct forms of growth are associated with different molecular profiles (e.g., mRNAs, non-coding RNAs, and proteins), and targeting differentially regulated genes has therapeutic potential for heart failure. The effects of exercise on the cardiac and circulating lipidomes in comparison to disease are unclear. Lipidomic profiling was performed on hearts and plasma of mice subjected to swim endurance training or a cardiac disease model (moderate or severe pressure overload). Several sphingolipid species and phospholipids containing omega-3/6 fatty acids were distinctly altered in heart and/or plasma with exercise versus pressure overload. A subset of lipids was validated in an independent mouse model with heart failure and atrial fibrillation. This study highlights the adaptations that occur to lipid profiles in response to endurance training versus pathology and provides a resource to investigate potential therapeutic targets and biomarkers.


Subject(s)
Biomarkers/blood , Phospholipids/blood , Animals , Atrial Fibrillation/blood , Atrial Fibrillation/metabolism , Cardiomegaly/blood , Cardiomegaly/metabolism , Disease Models, Animal , Heart Failure/blood , Heart Failure/metabolism , Humans , Mice , Myocardium/metabolism , Myocardium/pathology , Phospholipids/metabolism , Physical Conditioning, Animal , Sphingolipids/blood , Sphingolipids/metabolism
5.
Physiol Rev ; 98(1): 419-475, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29351515

ABSTRACT

The benefits of exercise on the heart are well recognized, and clinical studies have demonstrated that exercise is an intervention that can improve cardiac function in heart failure patients. This has led to significant research into understanding the key mechanisms responsible for exercise-induced cardiac protection. Here, we summarize molecular mechanisms that regulate exercise-induced cardiac myocyte growth and proliferation. We discuss in detail the effects of exercise on other cardiac cells, organelles, and systems that have received less or little attention and require further investigation. This includes cardiac excitation and contraction, mitochondrial adaptations, cellular stress responses to promote survival (heat shock response, ubiquitin-proteasome system, autophagy-lysosomal system, endoplasmic reticulum unfolded protein response, DNA damage response), extracellular matrix, inflammatory response, and organ-to-organ crosstalk. We summarize therapeutic strategies targeting known regulators of exercise-induced protection and the challenges translating findings from bench to bedside. We conclude that technological advancements that allow for in-depth profiling of the genome, transcriptome, proteome and metabolome, combined with animal and human studies, provide new opportunities for comprehensively defining the signaling and regulatory aspects of cell/organelle functions that underpin the protective properties of exercise. This is likely to lead to the identification of novel biomarkers and therapeutic targets for heart disease.


Subject(s)
Cardiovascular Physiological Phenomena , Exercise/physiology , Heart Diseases/prevention & control , Heart/physiology , Myocytes, Cardiac/physiology , Animals , Genome , Humans , Transcriptome
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(3): 219-234, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29217479

ABSTRACT

Cardiac myocyte membranes contain lipids which remodel dramatically in response to heart growth and remodeling. Lipid species have both structural and functional roles. Physiological and pathological cardiac remodeling have very distinct phenotypes, and the identification of molecular differences represent avenues for therapeutic interventions. Whether the abundance of specific lipid classes is different in physiological and pathological models was largely unknown. The aim of this study was to determine whether distinct lipids are regulated in settings of physiological and pathological remodeling, and if so, whether modulation of differentially regulated lipids could modulate heart size and function. Lipidomic profiling was performed on cardiac-specific transgenic mice with 1) physiological cardiac hypertrophy due to increased Insulin-like Growth Factor 1 (IGF1) receptor or Phosphoinositide 3-Kinase (PI3K) signaling, 2) small hearts due to depressed PI3K signaling (dnPI3K), and 3) failing hearts due to dilated cardiomyopathy (DCM). In hearts of dnPI3K and DCM mice, several phospholipids (plasmalogens) were decreased and sphingolipids increased compared to mice with physiological hypertrophy. To assess whether restoration of plasmalogens could restore heart size or cardiac function, dnPI3K and DCM mice were administered batyl alcohol (BA; precursor to plasmalogen biosynthesis) in the diet for 16weeks. BA supplementation increased a major plasmalogen species (p18:0) in the heart but had no effect on heart size or function. This may be due to the concurrent reduction in other plasmalogen species (p16:0 and p18:1) with BA. Here we show that lipid species are differentially regulated in settings of physiological and pathological remodeling. Restoration of lipid species in the failing heart warrants further examination.


Subject(s)
Cardiomegaly/metabolism , Glyceryl Ethers/pharmacology , Lipid Metabolism/drug effects , Myocardium/metabolism , Plasmalogens/metabolism , Ventricular Remodeling/drug effects , Animals , Cardiomegaly/drug therapy , Cardiomegaly/genetics , Cardiomegaly/pathology , Mice , Mice, Transgenic , Myocardium/pathology , Plasmalogens/genetics , Ventricular Remodeling/genetics
7.
Adv Exp Med Biol ; 1000: 187-210, 2017.
Article in English | MEDLINE | ID: mdl-29098623

ABSTRACT

Regular physical activity or exercise training can lead to heart enlargement known as cardiac hypertrophy. Cardiac hypertrophy is broadly defined as an increase in heart mass. In adults, cardiac hypertrophy is often considered a poor prognostic sign because it often progresses to heart failure. Heart enlargement in a setting of cardiac disease is referred to as pathological cardiac hypertrophy and is typically characterized by cell death and depressed cardiac function. By contrast, physiological cardiac hypertrophy, as occurs in response to chronic exercise training (i.e. the 'athlete's heart'), is associated with normal or enhanced cardiac function. The following chapter describes the morphologically distinct types of heart growth, and the key role of the insulin-like growth factor 1 (IGF1) - phosphoinositide 3-kinase (PI3K)-Akt signaling pathway in regulating exercise-induced physiological cardiac hypertrophy and cardiac protection. Finally we summarize therapeutic approaches that target the IGF1-PI3K-Akt signaling pathway which are showing promise in preclinical models of heart disease.


Subject(s)
Cardiomegaly/physiopathology , Exercise/physiology , Insulin-Like Growth Factor I/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Cardiomegaly/metabolism , Humans , Models, Cardiovascular , Physical Conditioning, Animal/physiology
9.
RNA Biol ; 14(5): 500-513, 2017 05 04.
Article in English | MEDLINE | ID: mdl-27124358

ABSTRACT

Expression of the miR-34 family (miR-34a, -34b, -34c) is elevated in settings of heart disease, and inhibition with antimiR-34a/antimiR-34 has emerged as a promising therapeutic strategy. Under chronic cardiac disease settings, targeting the entire miR-34 family is more effective than targeting miR-34a alone. The identification of transcription factor (TF)-miRNA regulatory networks has added complexity to understanding the therapeutic potential of miRNA-based therapies. Here, we sought to determine whether antimiR-34 targets secondary miRNAs via TFs which could contribute to antimiR-34-mediated protection. Using miRNA-Seq we identified differentially regulated miRNAs in hearts from mice with cardiac pathology due to transverse aortic constriction (TAC), and focused on miRNAs which were also regulated by antimiR-34. Two clusters of stress-responsive miRNAs were classified as "pathological" and "cardioprotective," respectively. Using ChIPBase we identified 45 TF binding sites on the promoters of "pathological" and "cardioprotective" miRNAs, and 5 represented direct targets of miR-34, with the capacity to regulate other miRNAs. Knockdown studies in a cardiomyoblast cell line demonstrated that expression of 2 "pathological" miRNAs (let-7e, miR-31) was regulated by one of the identified TFs. Furthermore, by qPCR we confirmed that expression of let-7e and miR-31 was lower in hearts from antimiR-34 treated TAC mice; this may explain why targeting the entire miR-34 family is more effective than targeting miR-34a alone. Finally, we showed that Acsl4 (a common target of miR-34, let-7e and miR-31) was increased in hearts from TAC antimiR-34 treated mice. In summary, antimiR-34 regulates the expression of other miRNAs and this has implications for drug development.


Subject(s)
Cardiomegaly/therapy , Gene Regulatory Networks , Heart Failure/therapy , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Adult , Analysis of Variance , Animals , Cardiomegaly/metabolism , Cell Line , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Disease Models, Animal , Gene Expression Regulation , Heart Failure/metabolism , Heart Ventricles/chemistry , Heart Ventricles/metabolism , Humans , Male , Mice , Mice, Inbred Strains , MicroRNAs/analysis , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/metabolism , Placebos , Sequence Analysis, RNA , Transcription Factors/genetics , Transcription Factors/metabolism
10.
J Physiol ; 594(20): 5959-5974, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27270487

ABSTRACT

KEY POINTS: MicroRNA (miRNA)-based therapies are in development for numerous diseases, including heart disease. Currently, very limited basic information is available on the regulation of specific miRNAs in male and female hearts in settings of disease. The identification of sex-specific miRNA signatures has implications for translation into the clinic and suggests the need for customised therapy. In the present study, we found that a miRNA-based treatment inhibiting miRNA-34a (miR-34a) was more effective in females in a setting of moderate dilated cardiomyopathy than in males. Furthermore, the treatment showed little benefit for either sex in a setting of more severe dilated cardiomyopathy associated with atrial fibrillation. The results highlight the importance of understanding the effect of miRNA-based therapies in cardiac disease settings in males and females. ABSTRACT: MicroRNA (miRNA)-34a (miR-34a) is elevated in the diseased heart in mice and humans. Previous studies have shown that inhibiting miR-34a in male mice in settings of pathological cardiac hypertrophy or ischaemia protects the heart against progression to heart failure. Whether inhibition of miR-34a protects the female heart is unknown. Furthermore, the therapeutic potential of silencing miR-34a in settings of dilated cardiomyopathy (DCM) and atrial fibrillation (AF) has not been assessed previously. In the present study, we examined the effect of silencing miR-34a in males and females in (1) a model of moderate DCM and (2) a model of severe DCM with AF. The cardiac disease models were administered with a locked nucleic acid-modified oligonucleotide (LNA-antimiR-34a) at 6-7 weeks of age when the models display cardiac dysfunction and conduction abnormalities. Cardiac function and morphology were measured 6 weeks after treatment. In the present study, we show that inhibition of miR-34a provides more protection in the DCM model in females than males. Disease prevention in LNA-antimiR-34a treated DCM female mice was characterized by attenuated heart enlargement and lung congestion, lower expression of cardiac stress genes (B-type natriuretic peptide, collagen gene expression), less cardiac fibrosis and better cardiac function. There was no evidence of significant protection in the severe DCM and AF model in either sex. Sex- and treatment-dependent regulation of miRNAs was also identified in the diseased heart, and may explain the differential response of males and females. These studies highlight the importance of examining the impact of miRNA-based drugs in both sexes and under different disease conditions.


Subject(s)
Cardiomegaly/metabolism , Cardiomyopathy, Dilated/metabolism , Heart Failure/metabolism , Heart/physiopathology , MicroRNAs/metabolism , Animals , Cardiomegaly/physiopathology , Disease Models, Animal , Female , Heart Failure/physiopathology , Male , Mice , Natriuretic Peptides/metabolism , Sex Characteristics , Ventricular Remodeling/physiology
11.
Sci Rep ; 6: 22442, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26928825

ABSTRACT

Expression of miR-154 is upregulated in the diseased heart and was previously shown to be upregulated in the lungs of patients with pulmonary fibrosis. However, the role of miR-154 in a model of sustained pressure overload-induced cardiac hypertrophy and fibrosis had not been assessed. To examine the role of miR-154 in the diseased heart, adult male mice were subjected to transverse aortic constriction for four weeks, and echocardiography was performed to confirm left ventricular hypertrophy and cardiac dysfunction. Mice were then subcutaneously administered a locked nucleic acid antimiR-154 or control over three consecutive days (25 mg/kg/day) and cardiac function was assessed 8 weeks later. Here, we demonstrate that therapeutic inhibition of miR-154 in mice with pathological hypertrophy was able to protect against cardiac dysfunction and attenuate adverse cardiac remodelling. The improved cardiac phenotype was associated with attenuation of heart and cardiomyocyte size, less cardiac fibrosis, lower expression of atrial and B-type natriuretic peptide genes, attenuation of profibrotic markers, and increased expression of p15 (a miR-154 target and cell cycle inhibitor). In summary, this study suggests that miR-154 may represent a novel target for the treatment of cardiac pathologies associated with cardiac fibrosis, hypertrophy and dysfunction.


Subject(s)
Hypertrophy, Left Ventricular/genetics , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Pulmonary Fibrosis/genetics , Ventricular Remodeling/genetics , Animals , Aorta/surgery , Atrial Natriuretic Factor/biosynthesis , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , Disease Models, Animal , Echocardiography , Hypertension/pathology , Hypertrophy, Left Ventricular/drug therapy , Hypertrophy, Left Ventricular/surgery , Male , Mice , Mice, Inbred C57BL , Natriuretic Peptide, Brain/biosynthesis , Oligonucleotides/genetics , Oligonucleotides/pharmacology , Ventricular Remodeling/drug effects
13.
Future Med Chem ; 7(13): 1771-92, 2015.
Article in English | MEDLINE | ID: mdl-26399457

ABSTRACT

miRNAs are small non-coding RNAs (ncRNAs), which regulate gene expression. Here, the authors describe the contribution of miRNAs to cardiac biology and disease. They discuss various strategies for manipulating miRNA activity including antisense oligonucleotides (antimiRs, blockmiRs), mimics, miRNA sponges, Tough Decoys and miRNA mowers. They review developments in chemistries (e.g., locked nucleic acid) and modifications (sugar, 'ZEN', peptide nucleic acids) and miRNA delivery tools (viral vectors, liposomes, nanoparticles, pHLIP). They summarize potential miRNA therapeutic targets for heart disease based on preclinical studies. Finally, the authors review current progress of miRNA therapeutics in clinical development for HCV and cancer, and discuss challenges that will need to be overcome for similar therapies to enter the clinic for patients with cardiac disease.


Subject(s)
Cardiovascular Diseases/drug therapy , Drug Discovery/methods , MicroRNAs/therapeutic use , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Gene Expression Regulation/drug effects , Heart/drug effects , Humans , MicroRNAs/chemistry , MicroRNAs/genetics , MicroRNAs/pharmacology , Myocardium/pathology
14.
Epigenetics ; 10(5): 418-30, 2015.
Article in English | MEDLINE | ID: mdl-25941940

ABSTRACT

Pharmacological histone deacetylase (HDAC) inhibitors attenuate pathological cardiac remodeling and hypertrophic gene expression; yet, the direct histone targets remain poorly characterized. Since the inhibition of HDAC activity is associated with suppressing hypertrophy, we hypothesized histone acetylation would target genes implicated in cardiac remodeling. Trichostatin A (TSA) regulates cardiac gene expression and attenuates transverse aortic constriction (TAC) induced hypertrophy. We used chromatin immunoprecipitation (ChIP) coupled with massive parallel sequencing (ChIP-seq) to map, for the first time, genome-wide histone acetylation changes in a preclinical model of pathological cardiac hypertrophy and attenuation of pathogenesis with TSA. Pressure overload-induced cardiac hypertrophy was associated with histone acetylation of genes implicated in cardiac contraction, collagen deposition, inflammation, and extracellular matrix identified by ChIP-seq. Gene set enrichment analysis identified NF-kappa B (NF-κB) transcription factor activation with load induced hypertrophy. Increased histone acetylation was observed on the promoters of NFκB target genes (Icam1, Vcam1, Il21r, Il6ra, Ticam2, Cxcl10) consistent with gene activation in the hypertrophied heart. Surprisingly, TSA attenuated pressure overload-induced cardiac hypertrophy and the suppression of NFκB target genes by broad histone deacetylation. Our results suggest a mechanism for cardioprotection subject to histone deacetylation as a previously unknown target, implicating the importance of inflammation by pharmacological HDAC inhibition. The results of this study provides a framework for HDAC inhibitor function in the heart and argues the long held views of acetylation is subject to more flexibility than previously thought.


Subject(s)
Acetylation/drug effects , Cardiomegaly/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Animals , Aorta/surgery , Cardiomegaly/genetics , Cardiomegaly/surgery , Gene Expression Regulation/drug effects , Histones/metabolism , Male , Mice, Inbred C57BL , Myocardium/metabolism , NF-kappa B/metabolism
15.
Arch Toxicol ; 89(9): 1401-38, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25708889

ABSTRACT

The onset of heart failure is typically preceded by cardiac hypertrophy, a response of the heart to increased workload, a cardiac insult such as a heart attack or genetic mutation. Cardiac hypertrophy is usually characterized by an increase in cardiomyocyte size and thickening of ventricular walls. Initially, such growth is an adaptive response to maintain cardiac function; however, in settings of sustained stress and as time progresses, these changes become maladaptive and the heart ultimately fails. In this review, we discuss the key features of pathological cardiac hypertrophy and the numerous mediators that have been found to be involved in the pathogenesis of cardiac hypertrophy affecting gene transcription, calcium handling, protein synthesis, metabolism, autophagy, oxidative stress and inflammation. We also discuss new mediators including signaling proteins, microRNAs, long noncoding RNAs and new findings related to the role of calcineurin and calcium-/calmodulin-dependent protein kinases. We also highlight mediators and processes which contribute to the transition from adaptive cardiac remodeling to maladaptive remodeling and heart failure. Treatment strategies for heart failure commonly include diuretics, angiotensin converting enzyme inhibitors, angiotensin II receptor blockers and ß-blockers; however, mortality rates remain high. Here, we discuss new therapeutic approaches (e.g., RNA-based therapies, dietary supplementation, small molecules) either entering clinical trials or in preclinical development. Finally, we address the challenges that remain in translating these discoveries to new and approved therapies for heart failure.


Subject(s)
Cardiomegaly/physiopathology , Heart Failure/physiopathology , Myocytes, Cardiac/pathology , Animals , Cardiomegaly/therapy , Heart Failure/therapy , Heart Ventricles/pathology , Humans , Signal Transduction/physiology
17.
Nat Commun ; 5: 5705, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25489988

ABSTRACT

Heart failure (HF) and atrial fibrillation (AF) share common risk factors, frequently coexist and are associated with high mortality. Treatment of HF with AF represents a major unmet need. Here we show that a small molecule, BGP-15, improves cardiac function and reduces arrhythmic episodes in two independent mouse models, which progressively develop HF and AF. In these models, BGP-15 treatment is associated with increased phosphorylation of the insulin-like growth factor 1 receptor (IGF1R), which is depressed in atrial tissue samples from patients with AF. Cardiac-specific IGF1R transgenic overexpression in mice with HF and AF recapitulates the protection observed with BGP-15. We further demonstrate that BGP-15 and IGF1R can provide protection independent of phosphoinositide 3-kinase-Akt and heat-shock protein 70; signalling mediators often defective in the aged and diseased heart. As BGP-15 is safe and well tolerated in humans, this study uncovers a potential therapeutic approach for HF and AF.


Subject(s)
Atrial Fibrillation/drug therapy , Heart Failure/prevention & control , Oximes/chemistry , Piperidines/chemistry , Animals , Caveolin 1/metabolism , Caveolin 3/metabolism , Disease Models, Animal , Electrocardiography , G(M3) Ganglioside/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Microarray Analysis , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Somatomedin/metabolism , Risk Factors , Signal Transduction , Transgenes
18.
FASEB J ; 28(12): 5097-110, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25145628

ABSTRACT

Expression of microRNA-652 (miR-652) increases in the diseased heart, decreases in a setting of cardioprotection, and is inversely correlated with heart function. The aim of this study was to assess the therapeutic potential of inhibiting miR-652 in a mouse model with established pathological hypertrophy and cardiac dysfunction due to pressure overload. Mice were subjected to a sham operation or transverse aortic constriction (TAC) for 4 wk to induce hypertrophy and cardiac dysfunction, followed by administration of a locked nucleic acid (LNA)-antimiR-652 (miR-652 inhibitor) or LNA control. Cardiac function was assessed before and 8 wk post-treatment. Expression of miR-652 increased in hearts subjected to TAC compared to sham surgery (2.9-fold), and this was suppressed by ∼95% in LNA-antimiR-652-treated TAC mice. Inhibition of miR-652 improved cardiac function in TAC mice (fractional shortening:29±1% at 4 wk post-TAC compared to 35±1% post-treatment) and attenuated cardiac hypertrophy. Improvement in heart function was associated with reduced cardiac fibrosis, less apoptosis and B-type natriuretic peptide gene expression, and preserved angiogenesis. Mechanistically, we identified Jagged1 (a Notch1 ligand) as a novel direct target of miR-652. In summary, these studies provide the first evidence that silencing of miR-652 protects the heart against pathological remodeling and improves heart function.


Subject(s)
Cardiomegaly/genetics , Gene Silencing , Heart/physiopathology , MicroRNAs/genetics , Animals , Cells, Cultured , Mice , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
19.
PLoS One ; 9(2): e90337, 2014.
Article in English | MEDLINE | ID: mdl-24587330

ABSTRACT

Therapeutic inhibition of the miR-34 family (miR-34a,-b,-c), or miR-34a alone, have emerged as promising strategies for the treatment of cardiac pathology. However, before advancing these approaches further for potential entry into the clinic, a more comprehensive assessment of the therapeutic potential of inhibiting miR-34a is required for two key reasons. First, miR-34a has ∼40% fewer predicted targets than the miR-34 family. Hence, in cardiac stress settings in which inhibition of miR-34a provides adequate protection, this approach is likely to result in less potential off-target effects. Secondly, silencing of miR-34a alone may be insufficient in settings of established cardiac pathology. We recently demonstrated that inhibition of the miR-34 family, but not miR-34a alone, provided benefit in a chronic model of myocardial infarction. Inhibition of miR-34 also attenuated cardiac remodeling and improved heart function following pressure overload, however, silencing of miR-34a alone was not examined. The aim of this study was to assess whether inhibition of miR-34a could attenuate cardiac remodeling in a mouse model with pre-existing pathological hypertrophy. Mice were subjected to pressure overload via constriction of the transverse aorta for four weeks and echocardiography was performed to confirm left ventricular hypertrophy and systolic dysfunction. After four weeks of pressure overload (before treatment), two distinct groups of animals became apparent: (1) mice with moderate pathology (fractional shortening decreased ∼20%) and (2) mice with severe pathology (fractional shortening decreased ∼37%). Mice were administered locked nucleic acid (LNA)-antimiR-34a or LNA-control with an eight week follow-up. Inhibition of miR-34a in mice with moderate cardiac pathology attenuated atrial enlargement and maintained cardiac function, but had no significant effect on fetal gene expression or cardiac fibrosis. Inhibition of miR-34a in mice with severe pathology provided no therapeutic benefit. Thus, therapies that inhibit miR-34a alone may have limited potential in settings of established cardiac pathology.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/physiopathology , Gene Silencing , MicroRNAs/genetics , Animals , Cardiomyopathy, Hypertrophic/pathology , Disease Models, Animal , Echocardiography , Fibrosis , Gene Expression Regulation , Male , Mice , Severity of Illness Index , Ventricular Remodeling/genetics
20.
Future Med Chem ; 6(2): 205-22, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24467244

ABSTRACT

Cardiac hypertrophy is broadly defined as an increase in heart mass. Heart enlargement in a setting of cardiac disease is referred to as pathological hypertrophy and often progresses to heart failure. Physiological hypertrophy refers to heart growth in response to postnatal development, exercise training and pregnancy, and is an adaptive response associated with the activation of cardioprotective signaling cascades. miRNAs have emerged as novel therapeutic targets for numerous pathologies, and miRNA-based therapies have already entered clinical trials. The identification of miRNAs differentially regulated during physiological growth may open up new therapeutic approaches for heart failure. In this review, we present information on miRNAs regulated in models of physiological hypertrophy, describe preclinical cardiac disease studies that have successfully targeted miRNAs regulated in settings of physiological growth (miR-34, miR-15, miR-199b, miR-208a and miR-378), and discuss challenges to overcome for the safe entry of miRNA-based therapies into the clinic for heart failure patients.


Subject(s)
Cardiomegaly/physiopathology , MicroRNAs/metabolism , Animals , Cardiomegaly/metabolism , Cardiomegaly/therapy , Female , Heart/growth & development , Heart/physiology , Hepatitis C/therapy , MicroRNAs/antagonists & inhibitors , MicroRNAs/therapeutic use , Physical Conditioning, Animal , Pregnancy , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...