Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Mol Ther Nucleic Acids ; 35(2): 102174, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38584818

ABSTRACT

Dystrophic cardiomyopathy is a significant feature of Duchenne muscular dystrophy (DMD). Increased cardiomyocyte cytosolic calcium (Ca2+) and interstitial fibrosis are major pathophysiological hallmarks that ultimately result in cardiac dysfunction. MicroRNA-25 (miR-25) has been identified as a suppressor of both sarcoplasmic reticulum calcium ATPase 2a (SERCA2a) and mothers against decapentaplegic homolog-7 (Smad7) proteins. In this study, we created a gene transfer using an miR-25 tough decoy (TuD) RNA inhibitor delivered via recombinant adeno-associated virus serotype 9 (AAV9) to evaluate the effect of miR-25 inhibition on cardiac and skeletal muscle function in aged dystrophin/utrophin haploinsufficient mice mdx/utrn (+/-), a validated transgenic murine model of DMD. We found that the intravenous delivery of AAV9 miR-25 TuD resulted in strong and stable inhibition of cardiac miR-25 levels, together with the restoration of SERCA2a and Smad7 expression. This was associated with the amelioration of cardiomyocyte interstitial fibrosis as well as recovered cardiac function. Furthermore, the direct quadricep intramuscular injection of AAV9 miR-25 TuD significantly restored skeletal muscle Smad7 expression, reduced tissue fibrosis, and enhanced skeletal muscle performance in mdx/utrn (+/-) mice. These results imply that miR-25 TuD gene transfer may be a novel therapeutic approach to restore cardiomyocyte Ca2+ homeostasis and abrogate tissue fibrosis in DMD.

2.
Int J Mol Sci ; 24(4)2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36834924

ABSTRACT

Impaired calcium uptake resulting from reduced expression and activity of the cardiac sarco-endoplasmic reticulum Ca2+ ATPase (SERCA2a) is a hallmark of heart failure (HF). Recently, new mechanisms of SERCA2a regulation, including post-translational modifications (PTMs), have emerged. Our latest analysis of SERCA2a PTMs has identified lysine acetylation as another PTM which might play a significant role in regulating SERCA2a activity. SERCA2a is acetylated, and that acetylation is more prominent in failing human hearts. In this study, we confirmed that p300 interacts with and acetylates SERCA2a in cardiac tissues. Several lysine residues in SERCA2a modulated by p300 were identified using in vitro acetylation assay. Analysis of in vitro acetylated SERCA2a revealed several lysine residues in SERCA2a susceptible to acetylation by p300. Among them, SERCA2a Lys514 (K514) was confirmed to be essential for SERCA2a activity and stability using an acetylated mimicking mutant. Finally, the reintroduction of an acetyl-mimicking mutant of SERCA2a (K514Q) into SERCA2 knockout cardiomyocytes resulted in deteriorated cardiomyocyte function. Taken together, our data demonstrated that p300-mediated acetylation of SERCA2a is a critical PTM that decreases the pump's function and contributes to cardiac impairment in HF. SERCA2a acetylation can be targeted for therapeutic aims for the treatment of HF.


Subject(s)
Heart Failure , Protein Processing, Post-Translational , Sarcoplasmic Reticulum Calcium-Transporting ATPases , p300-CBP Transcription Factors , Humans , Heart Failure/metabolism , Lysine/metabolism , Myocytes, Cardiac/metabolism , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
3.
Front Cardiovasc Med ; 9: 763544, 2022.
Article in English | MEDLINE | ID: mdl-35557546

ABSTRACT

Duchenne muscular dystrophy (DMD) is a genetic disorder characterized by progressive muscle degeneration due to dystrophin gene mutations. Patients with DMD initially experience muscle weakness in their limbs during adolescence. With age, patients develop fatal respiratory and cardiac dysfunctions. During the later stages of the disease, severe cardiac fibrosis occurs, compromising cardiac function. Previously, our research showed that the matricellular protein CCN5 has antifibrotic properties. Therefore, we hypothesized that CCN5 gene transfer would ameliorate cardiac fibrosis and thus improve cardiac function in DMD-induced cardiomyopathy. We utilized mdx/utrn (±) haploinsufficient mice that recapitulated the DMD-disease phenotypes and used an adeno-associated virus serotype-9 viral vector for CCN5 gene transfer. We evaluated the onset of cardiac dysfunction using echocardiography and determined the experimental starting point in 13-month-old mice. Two months after CCN5 gene transfer, cardiac function was significantly enhanced, and cardiac fibrosis was ameliorated. Additionally, running performance was improved in CCN5 gene-transfected mice. Furthermore, in silico gene profiling analysis identified utrophin as a novel transcriptional target of CCN5. This was supplemented by a utrophin promoter assay and RNA-seq analysis, which confirmed that CCN5 was directly associated with utrophin expression. Our results showed that CCN5 may be a promising therapeutic molecule for DMD-induced cardiac and skeletal dysfunction.

4.
Cardiovasc Res ; 118(15): 3140-3150, 2022 12 09.
Article in English | MEDLINE | ID: mdl-35191471

ABSTRACT

AIMS: A mutation in the phospholamban (PLN) gene, leading to deletion of Arg14 (R14del), has been associated with malignant arrhythmias and ventricular dilation. Identifying pre-symptomatic carriers with vulnerable myocardium is crucial because arrhythmia can result in sudden cardiac death, especially in young adults with PLN-R14del mutation. This study aimed at assessing the efficiency and efficacy of in vivo genome editing, using CRISPR/Cas9 and a cardiotropic adeno-associated virus-9 (AAV9), in improving cardiac function in young adult mice expressing the human PLN-R14del. METHODS AND RESULTS: Humanized mice were generated expressing human wild-type (hPLN-WT) or mutant (hPLN-R14del) PLN in the heterozygous state, mimicking human carriers. Cardiac magnetic resonance imaging at 12 weeks of age showed bi-ventricular dilation and increased stroke volume in mutant vs. WT mice, with no deficit in ejection fraction or cardiac output. Challenge of ex vivo hearts with isoproterenol and rapid pacing unmasked higher propensity for sustained ventricular tachycardia (VT) in hPLN-R14del relative to hPLN-WT. Specifically, the VT threshold was significantly reduced (20.3 ± 1.2 Hz in hPLN-R14del vs. 25.7 ± 1.3 Hz in WT, P < 0.01) reflecting higher arrhythmia burden. To inactivate the R14del allele, mice were tail-vein-injected with AAV9.CRISPR/Cas9/gRNA or AAV9 empty capsid (controls). CRISPR-Cas9 efficiency was evaluated by droplet digital polymerase chain reaction and NGS-based amplicon sequencing. In vivo gene editing significantly reduced end-diastolic and stroke volumes in hPLN-R14del CRISPR-treated mice compared to controls. Susceptibility to VT was also reduced, as the VT threshold was significantly increased relative to controls (30.9 ± 2.3 Hz vs. 21.3 ± 1.5 Hz; P < 0.01). CONCLUSIONS: This study is the first to show that disruption of hPLN-R14del allele by AAV9-CRISPR/Cas9 improves cardiac function and reduces VT susceptibility in humanized PLN-R14del mice, offering preclinical evidence for translatable approaches to therapeutically suppress the arrhythmogenic phenotype in human patients with PLN-R14del disease.


Subject(s)
Cardiomyopathies , Gene Editing , Humans , Mice , Animals , Cardiomyopathies/genetics , Cardiomyopathies/therapy
5.
J Cell Mol Med ; 24(13): 7214-7227, 2020 07.
Article in English | MEDLINE | ID: mdl-32485073

ABSTRACT

Extracellular vesicles (EVs) have recently emerged as an important carrier for various genetic materials including microRNAs (miRs). Growing evidences suggested that several miRs transported by EVs were particularly involved in modulating cardiac function. However, it has remained unclear what miRs are enriched in EVs and play an important role in the pathological condition. Therefore, we established the miR expression profiles in EVs from murine normal and failing hearts and consecutively identified substantially altered miRs. In addition, we have performed bioinformatics approach to predict potential cardiac outcomes through the identification of miR targets. Conclusively, we observed approximately 63% of predicted targets were validated with previous reports. Notably, the predicted targets by this approach were often involved in both beneficial and malicious signalling pathways, which may reflect heterogeneous cellular origins of EVs in tissues. Lastly, there has been an active debate on U6 whether it is a proper control. Through further analysis of EV miR profiles, miR-676 was identified as a superior reference control due to its consistent and abundant expressions. In summary, our results contribute to identifying specific EV miRs for the potential therapeutic targets in heart failure and suggest that miR-676 as a new reference control for the EV miR studies.


Subject(s)
Extracellular Vesicles/genetics , Gene Expression Profiling , Heart Failure/genetics , MicroRNAs/genetics , Animals , Down-Regulation/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Proteomics , Reproducibility of Results , Up-Regulation/genetics
6.
J Vis Exp ; (155)2020 01 17.
Article in English | MEDLINE | ID: mdl-32009647

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) provide a valuable human source for studying the basic science of calcium (Ca2+) handling and signaling pathways as well as high-throughput drug screening and toxicity assays. Herein, we provide a detailed description of the methodologies used to generate high-quality iPSC-CMs that can consistently reproduce molecular and functional characteristics across different cell lines. Additionally, a method is described to reliably assess their functional characterization through the evaluation of Ca2+ handling properties. Low oxygen (O2) conditions, lactate selection, and prolonged time in culture produce high-purity and high-quality ventricular-like cardiomyocytes. Similar to isolated adult rat cardiomyocytes (ARCMs), 3-month-old iPSC-CMs exhibit higher Ca2+ amplitude, faster rate of Ca2+ reuptake (decay-tau), and a positive lusitropic response to ß-adrenergic stimulation compared to day 30 iPSC-CMs. The strategy is technically simple, cost-effective, and reproducible. It provides a robust platform to model cardiac disease and for the large-scale drug screening to target Ca2+ handling proteins.


Subject(s)
Calcium/metabolism , Heart Ventricles/cytology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Cell Differentiation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Rats, Sprague-Dawley , Time Factors
7.
Circulation ; 141(15): 1249-1265, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32078387

ABSTRACT

BACKGROUND: The adult mammalian heart has limited regenerative capacity, mostly attributable to postnatal cardiomyocyte cell cycle arrest. In the last 2 decades, numerous studies have explored cardiomyocyte cell cycle regulatory mechanisms to enhance myocardial regeneration after myocardial infarction. Pkm2 (Pyruvate kinase muscle isoenzyme 2) is an isoenzyme of the glycolytic enzyme pyruvate kinase. The role of Pkm2 in cardiomyocyte proliferation, heart development, and cardiac regeneration is unknown. METHODS: We investigated the effect of Pkm2 in cardiomyocytes through models of loss (cardiomyocyte-specific Pkm2 deletion during cardiac development) or gain using cardiomyocyte-specific Pkm2 modified mRNA to evaluate Pkm2 function and regenerative affects after acute or chronic myocardial infarction in mice. RESULTS: Here, we identify Pkm2 as an important regulator of the cardiomyocyte cell cycle. We show that Pkm2 is expressed in cardiomyocytes during development and immediately after birth but not during adulthood. Loss of function studies show that cardiomyocyte-specific Pkm2 deletion during cardiac development resulted in significantly reduced cardiomyocyte cell cycle, cardiomyocyte numbers, and myocardial size. In addition, using cardiomyocyte-specific Pkm2 modified RNA, our novel cardiomyocyte-targeted strategy, after acute or chronic myocardial infarction, resulted in increased cardiomyocyte cell division, enhanced cardiac function, and improved long-term survival. We mechanistically show that Pkm2 regulates the cardiomyocyte cell cycle and reduces oxidative stress damage through anabolic pathways and ß-catenin. CONCLUSIONS: We demonstrate that Pkm2 is an important intrinsic regulator of the cardiomyocyte cell cycle and oxidative stress, and highlight its therapeutic potential using cardiomyocyte-specific Pkm2 modified RNA as a gene delivery platform.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle/physiology , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Regeneration/physiology , Thyroid Hormones/metabolism , Animals , Humans , Mice , Transfection , Thyroid Hormone-Binding Proteins
8.
Cell Death Dis ; 10(7): 511, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31263105

ABSTRACT

In ischemic human hearts, the induction of adenosine receptor A2B (ADORA2B) is associated with cardioprotection against ischemic heart damage, but the mechanism underlying this association remains unclear. Apaf-1-interacting protein (APIP) and ADORA2B transcript levels in human hearts are substantially higher in patients with heart failure than in controls. Interestingly, the APIP and ADORA2B mRNA levels are highly correlated with each other (R = 0.912). APIP expression was significantly increased in primary neonatal cardiomyocytes under hypoxic conditions and this induction reduced myocardial cell death via the activation of the AKT-HIF1α pathway. Accordingly, infarct sizes of APIP transgenic mice after left anterior descending artery ligation were significantly reduced compared to those of wild-type mice. Strikingly, knockdown of APIP expression impaired the cytoprotective effects of ADORA2B during hypoxic damage. Immunoprecipitation and proximity ligation assays revealed that APIP interacts with ADORA2B, leading to the stabilization of both proteins by interfering with lysosomal degradation, and to the activation of the downstream PKA-CREB signaling pathways. ADORA2B levels in the hearts of APIPTg/Tg, APIPTg/+, and Apip+/- mice were proportionally downregulated. In addition, ADORA2B D296G derived from the rs200741295 polymorphism failed to bind to APIP and did not exert cardioprotective activity during hypoxia. Moreover, Adora2b D296G knock-in mice were more vulnerable than control mice to myocardial infarction and intentional increases in APIP levels overcame the defective protection of the ADORA2B SNP against ischemic injury. Collectively, APIP is crucial for cardioprotection against myocardial infarction by virtue of binding to and stabilizing ADORA2B, thereby dampening ischemic heart injury.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Receptor, Adenosine A2B/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Cell Line , Cells, Cultured , Female , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Polymorphism, Genetic/genetics , Polymorphism, Single Nucleotide/genetics , Receptor, Adenosine A2B/genetics , Signal Transduction/genetics , Signal Transduction/physiology
10.
J Mol Cell Cardiol ; 129: 58-68, 2019 04.
Article in English | MEDLINE | ID: mdl-30771307

ABSTRACT

The reduced expression of cardiac sarco-endoplasmic reticulum Ca2+ ATPase (SERCA2a) is a hallmark of heart failure. We previously showed that miR-25 is a crucial transcriptional regulator of SERCA2a in the heart. However, the precise mechanism of cardiac miR-25 regulation is largely unknown. Literatures suggested that miR-25 is regulated by the transcriptional co-factor, sine oculis homeobox homolog 1 (Six1), which in turn is epigenetically regulated by polycomb repressive complex 2 (PRC 2) in cardiac progenitor cells. Therefore, we aimed to investigate whether Six1 and PRC2 are indeed involved in the regulation of the miR-25 level in the setting of heart failure. Six1 was up-regulated in the failing hearts of humans and mice. Overexpression of Six1 led to adverse cardiac remodeling, whereas knock-down of Six1 attenuated pressure overload-induced cardiac dysfunction. The adverse effects of Six1 were ameliorated by knock-down of miR-25. The epigenetic repression on the Six1 promoter by PRC2 was significantly reduced in failing hearts. Epigenetic repression of Six1 is relieved through a reduction of PRC2 activity in heart failure. Six1 up-regulates miR-25, which is followed by reduction of cardiac SERCA2a expression. Collectively, these data showed that the PRC2-Six1-miR-25 signaling axis is involved in heart failure. Our finding introduces new insight into potential treatments of heart failure.


Subject(s)
Heart Failure/genetics , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Polycomb Repressive Complex 2/metabolism , Signal Transduction , Animals , Epigenesis, Genetic , Gene Knockdown Techniques , Heart Failure/physiopathology , Homeodomain Proteins/genetics , Humans , Mice, Inbred C57BL , MicroRNAs/genetics , Pressure , Promoter Regions, Genetic , Up-Regulation/genetics , Ventricular Remodeling/genetics
11.
Circ Res ; 124(9): e63-e80, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30786847

ABSTRACT

RATIONALE: SERCA2a, sarco-endoplasmic reticulum Ca2+-ATPase, is a critical determinant of cardiac function. Reduced level and activity of SERCA2a are major features of heart failure. Accordingly, intensive efforts have been made to develop efficient modalities for SERCA2a activation. We showed that the activity of SERCA2a is enhanced by post-translational modification with SUMO1 (small ubiquitin-like modifier 1). However, the roles of other post-translational modifications on SERCA2a are still unknown. OBJECTIVE: In this study, we aim to assess the role of lysine acetylation on SERCA2a function and determine whether inhibition of lysine acetylation can improve cardiac function in the setting of heart failure. METHODS AND RESULTS: The acetylation of SERCA2a was significantly increased in failing hearts of humans, mice, and pigs, which is associated with the reduced level of SIRT1 (sirtuin 1), a class III histone deacetylase. Downregulation of SIRT1 increased the SERCA2a acetylation, which in turn led to SERCA2a dysfunction and cardiac defects at baseline. In contrast, pharmacological activation of SIRT1 reduced the SERCA2a acetylation, which was accompanied by recovery of SERCA2a function and cardiac defects in failing hearts. Lysine 492 (K492) was of critical importance for the regulation of SERCA2a activity via acetylation. Acetylation at K492 significantly reduced the SERCA2a activity, presumably through interfering with the binding of ATP to SERCA2a. In failing hearts, acetylation at K492 appeared to be mediated by p300 (histone acetyltransferase p300), a histone acetyltransferase. CONCLUSIONS: These results indicate that acetylation/deacetylation at K492, which is regulated by SIRT1 and p300, is critical for the regulation of SERCA2a activity in hearts. Pharmacological activation of SIRT1 can restore SERCA2a activity through deacetylation at K492. These findings might provide a novel strategy for the treatment of heart failure.


Subject(s)
Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sirtuin 1/metabolism , Acetylation , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cells, Cultured , E1A-Associated p300 Protein/metabolism , Heart Failure/enzymology , Heart Failure/genetics , Humans , Lysine/genetics , Lysine/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/pathology , Protein Processing, Post-Translational , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sirtuin 1/genetics , Swine
12.
Heart Fail Rev ; 24(4): 601-615, 2019 07.
Article in English | MEDLINE | ID: mdl-30666533

ABSTRACT

Experimental models of cardiac disease play a key role in understanding the pathophysiology of the disease and developing new therapies. The features of the experimental models should reflect the clinical phenotype, which can have a wide spectrum of underlying mechanisms. We review characteristics of commonly used experimental models of cardiac physiology and pathophysiology in all translational steps including in vitro, small animal, and large animal models. Understanding their characteristics and relevance to clinical disease is the key for successful translation to effective therapies.


Subject(s)
Heart Diseases/physiopathology , Heart/physiology , Models, Biological , Animals , Cell Line , Disease Models, Animal , Heart Diseases/etiology , Heart Diseases/pathology , Humans , In Vitro Techniques/methods , Myocytes, Cardiac/physiology , Tissue Engineering/methods
13.
Discov Med ; 28(155): 229-235, 2019.
Article in English | MEDLINE | ID: mdl-32053763

ABSTRACT

There has been significant progress in the field of gene therapy toward clinical application in recent years. Cardiac gene therapy has followed this trend, but at a slower pace compared to therapies targeting retinal, spinal, and blood disorders. New approaches targeting monogenic disorders are being developed at a rapid pace and studies in large animal models show promise. Meanwhile, several clinical trials are underway to prove the efficacy of gene therapy for cardiovascular diseases such as heart failure. In this concise review, we attempt to summarize recent highlights in technological advancements, preclinical studies, and clinical trials of cardiac gene therapy.


Subject(s)
Cardiovascular Diseases/genetics , Cardiovascular Diseases/therapy , Genetic Therapy/trends , Animals , Clinical Trials as Topic , Dependovirus/genetics , Gene Editing , Humans
14.
Circ Res ; 123(6): 673-685, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30355233

ABSTRACT

RATIONALE: Abnormal SUMOylation has emerged as a characteristic of heart failure (HF) pathology. Previously, we found reduced SUMO1 (small ubiquitin-like modifier 1) expression and SERCA2a (sarcoplasmic reticulum Ca2+-ATPase) SUMOylation in human and animal HF models. SUMO1 gene delivery or small molecule activation of SUMOylation restored SERCA2a SUMOylation and cardiac function in HF models. Despite the critical role of SUMO1 in HF, the regulatory mechanisms underlying SUMO1 expression are largely unknown. OBJECTIVE: To examine miR-146a-mediated SUMO1 regulation and its consequent effects on cardiac morphology and function. METHODS AND RESULTS: In this study, miR-146a was identified as a SUMO1-targeting microRNA in the heart. A strong correlation was observed between miR-146a and SUMO1 expression in failing mouse and human hearts. miR-146a was manipulated in cardiomyocytes through AAV9 (adeno-associated virus serotype 9)-mediated gene delivery, and cardiac morphology and function were analyzed by echocardiography and hemodynamics. Overexpression of miR-146a reduced SUMO1 expression, SERCA2a SUMOylation, and cardiac contractility in vitro and in vivo. The effects of miR-146a inhibition on HF pathophysiology were examined by transducing a tough decoy of miR-146a into mice subjected to transverse aortic constriction. miR-146a inhibition improved cardiac contractile function and normalized SUMO1 expression. The regulatory mechanisms of miR-146a upregulation were elucidated by examining the major miR-146a-producing cell types and transfer mechanisms. Notably, transdifferentiation of fibroblasts triggered miR-146a overexpression and secretion through extracellular vesicles, and the extracellular vesicle-associated miR-146a transfer was identified as the causative mechanism of miR-146a upregulation in failing cardiomyocytes. Finally, extracellular vesicles isolated from failing hearts were shown to contain high levels of miR-146a and exerted negative effects on the SUMO1/SERCA2a signaling axis and hence cardiomyocyte contractility. CONCLUSIONS: Taken together, our results show that miR-146a is a novel regulator of the SUMOylation machinery in the heart, which can be targeted for therapeutic intervention.


Subject(s)
Cardiomegaly/metabolism , Heart Failure/metabolism , MicroRNAs/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , SUMO-1 Protein/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Communication , Cell Transdifferentiation , Cells, Cultured , Disease Models, Animal , Down-Regulation , Fibroblasts/metabolism , Fibroblasts/pathology , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Mice , MicroRNAs/genetics , Myocytes, Cardiac/pathology , SUMO-1 Protein/genetics , Signal Transduction , Sumoylation
15.
Methods Mol Biol ; 1816: 3-14, 2018.
Article in English | MEDLINE | ID: mdl-29987807

ABSTRACT

Cardiovascular disease is one of the most common causes of deaths in clinics. Experimental models of cardiovascular diseases are essential to understand disease mechanism, to provide accurate diagnoses, and to develop new therapies. Large numbers of experimental models have been proposed and replicated by many laboratories in the past. Models with significant advantages are chosen and became more popular. Particularly, feasibility, reproducibility, and human disease resemblance are the common key factors for frequently used cardiovascular disease models. In this chapter, we provide a brief overview of these experimental models used for in vitro, in vivo, and in silico studies of cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/pathology , Cardiovascular System/pathology , Disease Models, Animal , Myocytes, Cardiac/pathology , Animals , Cardiovascular Diseases/physiopathology , Cardiovascular System/physiopathology , Computer Simulation , Humans , Models, Cardiovascular
16.
Methods Mol Biol ; 1816: 93-104, 2018.
Article in English | MEDLINE | ID: mdl-29987813

ABSTRACT

In vitro measurements of cardiomyocyte contractility and Ca2+ handling have been used as a platform for determining physiological consequence of various genetic manipulations and identifying potential therapeutic targets for the treatment of heart failure. The Myocyte Calcium and Contractility System (IonOptix) offers a simultaneous trace of sarcomere movements and changes of intracellular Ca2+ levels in a single cardiomyocyte. Herein, we describe a modified protocol for the isolation of adult cardiomyocytes from murine hearts and provide a step-by-step description on how to analyze cardiomyocyte Ca2+ transient and contractility data collected using the IonOptix system. In our modified protocol, we recommend a novel cannulation technique which simplifies this difficult method and leads to improved viability of isolated cardiomyocytes. In addition, a comprehensive analysis of intracellular Ca2+ handling, SR Ca2+ load, myofilament Ca2+ sensitivity, and cardiomyocyte contractility is described in order to provide important insights into myocardial mechanics.


Subject(s)
Calcium/metabolism , Myocardial Contraction , Myocytes, Cardiac/cytology , Animals , Calcium Signaling , Cardiac Catheterization/methods , Cell Culture Techniques/methods , Cell Separation/methods , Cells, Cultured , Mice , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Sarcomeres/metabolism , Sarcoplasmic Reticulum/metabolism
18.
J Am Coll Cardiol ; 70(14): 1744-1756, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28958332

ABSTRACT

BACKGROUND: Increased protein phosphatase-1 in heart failure (HF) induces molecular changes deleterious to the cardiac cell. Inhibiting protein phosphatase-1 through the overexpression of a constitutively active inhibitor-1 (I-1c) has been shown to reverse cardiac dysfunction in a model of ischemic HF. OBJECTIVES: This study sought to determine the therapeutic efficacy of a re-engineered adenoassociated viral vector carrying I-1c (BNP116.I-1c) in a preclinical model of nonischemic HF, and to assess thoroughly the safety of BNP116.I-1c gene therapy. METHODS: Volume-overload HF was created in Yorkshire swine by inducing severe mitral regurgitation. One month after mitral regurgitation induction, pigs were randomized to intracoronary delivery of either BNP116.I-1c (n = 6) or saline (n = 7). Therapeutic efficacy and safety were evaluated 2 months after gene delivery. Additionally, 24 naive pigs received different doses of BNP116.I-1c for safety evaluation. RESULTS: At 1 month after mitral regurgitation induction, pigs developed HF as evidenced by increased left ventricular end-diastolic pressure and left ventricular volume indexes. Treatment with BNP116.I-1c resulted in improved left ventricular ejection fraction (-5.9 ± 4.2% vs. 5.5 ± 4.0%; p < 0.001) and adjusted dP/dt maximum (-3.39 ± 2.44 s-1 vs. 1.30 ± 2.39 s-1; p = 0.007). Moreover, BNP116.I-1c-treated pigs also exhibited a significant increase in left atrial ejection fraction at 2 months after gene delivery (-4.3 ± 3.1% vs. 7.5 ± 3.1%; p = 0.02). In vitro I-1c gene transfer in isolated left atrial myocytes from both pigs and rats increased calcium transient amplitude, consistent with its positive impact on left atrial contraction. We found no evidence of adverse electrical remodeling, arrhythmogenicity, activation of a cellular immune response, or off-target organ damage by BNP116.I-1c gene therapy in pigs. CONCLUSIONS: Intracoronary delivery of BNP116.I-1c was safe and improved contractility of the left ventricle and atrium in a large animal model of nonischemic HF.


Subject(s)
Heart Failure/drug therapy , Heart Failure/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Protein Phosphatase 1 , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Delivery Systems , Drug Monitoring/methods , Enzyme Inhibitors/pharmacology , Genetic Therapy/methods , Genetic Vectors/pharmacology , Heart Failure/etiology , Heart Failure/physiopathology , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Swine , Treatment Outcome
19.
PLoS One ; 11(10): e0165569, 2016.
Article in English | MEDLINE | ID: mdl-27792751

ABSTRACT

Neointimal growth in the injured vasculature is largely facilitated by the proliferation of vascular smooth muscle cells (VSMC), which associates with reduced sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) activity. The gene transfer-mediated restoration of the SERCA2a level thus attenuates neointimal growth and VSMC proliferation. We previously reported that a peptide targeted to protein phosphatase 1, ψPLB-SE, normalizes SERCA2a activity in cardiomyocytes. In this study, we found that ψPLB-SE attenuated neointimal growth in balloon-injured rat carotid arteries, and the proliferation and migration of VSMC cultured in high-serum media (synthetic conditions). In parallel, ψPLB-SE inhibited the degradation of SERCA2a in the injured carotid arteries and VSMC under synthetic conditions. The calpain inhibitor MDL28170 also attenuated SERCA2a degradation and VSMC proliferation under synthetic conditions, indicating that calpain degrades SERCA2a. The Ca2+ ionophore A23187 induced SERCA2a degradation in VSMC, which was blocked by either ψPLB-SE or MDL28170. Additionally, ψPLB-SE normalized the cytosolic Ca2+ level in VSMC that was increased by either A23187 or synthetic stimulation. Collectively, these data indicate that ψPLB-SE corrects the abnormal Ca2+ handling by activating SERCA2a, which further protects SERCA2a from calpain-dependent degradation in VSMC. We conclude that ψPLB-SE may form the basis of a therapeutic strategy for vascular proliferative disorders.


Subject(s)
Calcium-Binding Proteins/pharmacology , Muscle, Smooth, Vascular/cytology , Peptide Fragments/pharmacology , Protein Phosphatase 1/metabolism , Proteolysis/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Calpain/metabolism , Cell Proliferation/drug effects , Humans , Male , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley
20.
J Mol Cell Cardiol ; 97: 278-85, 2016 08.
Article in English | MEDLINE | ID: mdl-27266388

ABSTRACT

Definitively identifying the cell type of newly generated cells in the heart and defining their origins are central questions in cardiac regenerative medicine. Currently, it is challenging to ascertain the myocardial identity and to track myocardial progeny during heart development and disease due to lack of proper genetic tools. This may lead to many misinterpretations of the findings in cardiac regenerative biology. In this study, we developed a set of novel mouse models by inserting double reporter genes nlacZ/H2B-GFP, mGFP/H2B-mCherry into the start codon of Tnnt2 and Myh6. nlacZ (nuclear lacZ) and mGFP (membrane GFP) are flanked by two LoxP sites in these animals. We found that the reporter genes faithfully recapitulated Tnnt2 and Myh6 cardiac expression from embryonic stage and adulthood. The reporter mice provide unprecedented robustness and fidelity for visualizing and tracing cardiomyocytes with nuclear or cell membrane localization signals. These animal models offer superior genetic tools to meet a critical need in studies of heart development, cardiac stem cell biology and cardiac regenerative medicine.


Subject(s)
Genetic Markers , Myocytes, Cardiac/metabolism , Phenotype , Animals , Cell Lineage/genetics , Female , Fluorescent Antibody Technique , Gene Expression , Genes, Reporter , Male , Mice , Mice, Transgenic , Myocardium , Myosin Heavy Chains/genetics , Organ Specificity/genetics , Recombinant Fusion Proteins , Regeneration , Regenerative Medicine , Troponin T/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...