Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Nat Cardiovasc Res ; 2(5): 467-485, 2023 May.
Article in English | MEDLINE | ID: mdl-37693816

ABSTRACT

The pleiotropic benefits of statins in cardiovascular diseases that are independent of their lipid-lowering effects have been well documented, but the underlying mechanisms remain elusive. Here we show that simvastatin significantly improves human induced pluripotent stem cell-derived endothelial cell functions in both baseline and diabetic conditions by reducing chromatin accessibility at transcriptional enhanced associate domain elements and ultimately at endothelial-to-mesenchymal transition (EndMT)-regulating genes in a yes-associated protein (YAP)-dependent manner. Inhibition of geranylgeranyltransferase (GGTase) I, a mevalonate pathway intermediate, repressed YAP nuclear translocation and YAP activity via RhoA signaling antagonism. We further identified a previously undescribed SOX9 enhancer downstream of statin-YAP signaling that promotes the EndMT process. Thus, inhibition of any component of the GGTase-RhoA-YAP-SRY box transcription factor 9 (SOX9) signaling axis was shown to rescue EndMT-associated endothelial dysfunction both in vitro and in vivo, especially under diabetic conditions. Overall, our study reveals an epigenetic modulatory role for simvastatin in repressing EndMT to confer protection against endothelial dysfunction.

2.
Stem Cell Res ; 65: 102969, 2022 12.
Article in English | MEDLINE | ID: mdl-36427473

ABSTRACT

Hispanics are the fastest-growing minority group in the United States. There has been a burgeoning interest in understanding the reasons underlying health disparities among this population. To facilitate the modeling and investigation of diseases that differentially impact Hispanics, we generated three induced pluripotent stem cell (iPSC) lines from the peripheral blood mononuclear cells (PBMCs) of healthy Hispanic subjects. All three lines exhibited normal morphology and karyotypes, robust expression of pluripotency markers, and the capacity for trilineage differentiation. The derivatives of these lines will serve as valuable ethnic-appropriate cell sources for further mechanistic studies on diseases impacting Hispanics.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Leukocytes, Mononuclear
3.
Sci Rep ; 12(1): 17605, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266453

ABSTRACT

Many cell-based therapies are challenged by the poor localization of introduced cells and the use of biomaterial scaffolds with questionable biocompatibility or bio-functionality. Endothelial progenitor cells (EPCs), a popular cell type used in cell-based therapies due to their robust angiogenic potential, are limited in their therapeutic capacity to develop into mature vasculature. Here, we demonstrate a joint delivery of human-derived endothelial progenitor cells (EPC) and smooth muscle cells (SMC) as a scaffold-free, bi-level cell sheet platform to improve ventricular remodeling and function in an athymic rat model of myocardial infarction. The transplanted bi-level cell sheet on the ischemic heart provides a biomimetic microenvironment and improved cell-cell communication, enhancing cell engraftment and angiogenesis, thereby improving ventricular remodeling. Notably, the increased density of vessel-like structures and upregulation of biological adhesion and vasculature developmental genes, such as Cxcl12 and Notch3, particularly in the ischemic border zone myocardium, were observed following cell sheet transplantation. We provide compelling evidence that this SMC-EPC bi-level cell sheet construct can be a promising therapy to repair ischemic cardiomyopathy.


Subject(s)
Myocardial Infarction , Ventricular Remodeling , Animals , Humans , Rats , Biocompatible Materials , Cells, Cultured , Myocardial Infarction/metabolism , Neovascularization, Physiologic , Stem Cell Transplantation , Stem Cells/metabolism
4.
Front Cardiovasc Med ; 9: 895291, 2022.
Article in English | MEDLINE | ID: mdl-35979018

ABSTRACT

Despite being responsible for half of heart failure-related hospitalizations, heart failure with preserved ejection fraction (HFpEF) has limited evidence-based treatment options. Currently, a substantial clinical issue is that the disease etiology is very heterogenous with no patient-specific treatment options. Modeling can provide a framework for evaluating alternative treatment strategies. Counterpulsation strategies have the capacity to improve left ventricular diastolic filling by reducing systolic blood pressure and augmenting the diastolic pressure that drives coronary perfusion. Here, we propose a framework for testing the effectiveness of a soft robotic extra-aortic counterpulsation strategy using a patient-specific closed-loop hemodynamic lumped parameter model of a patient with HFpEF. The soft robotic device prototype was characterized experimentally in a physiologically pressurized (50-150 mmHg) soft silicone vessel and modeled as a combination of a pressure source and a capacitance. The patient-specific model was created using open-source software and validated against hemodynamics obtained by imaging of a patient (male, 87 years, HR = 60 bpm) with HFpEF. The impact of actuation timing on the flows and pressures as well as systolic function was analyzed. Good agreement between the patient-specific model and patient data was achieved with relative errors below 5% in all categories except for the diastolic aortic root pressure and the end systolic volume. The most effective reduction in systolic pressure compared to baseline (147 vs. 141 mmHg) was achieved when actuating 350 ms before systole. In this case, flow splits were preserved, and cardiac output was increased (5.17 vs. 5.34 L/min), resulting in increased blood flow to the coronaries (0.15 vs. 0.16 L/min). Both arterial elastance (0.77 vs. 0.74 mmHg/mL) and stroke work (11.8 vs. 10.6 kJ) were decreased compared to baseline, however left atrial pressure increased (11.2 vs. 11.5 mmHg). A higher actuation pressure is associated with higher systolic pressure reduction and slightly higher coronary flow. The soft robotic device prototype achieves reduced systolic pressure, reduced stroke work, slightly increased coronary perfusion, but increased left atrial pressures in HFpEF patients. In future work, the framework could include additional physiological mechanisms, a larger patient cohort with HFpEF, and testing against clinically used devices.

7.
Cell ; 185(10): 1676-1693.e23, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35489334

ABSTRACT

Epidemiological studies reveal that marijuana increases the risk of cardiovascular disease (CVD); however, little is known about the mechanism. Δ9-tetrahydrocannabinol (Δ9-THC), the psychoactive component of marijuana, binds to cannabinoid receptor 1 (CB1/CNR1) in the vasculature and is implicated in CVD. A UK Biobank analysis found that cannabis was an risk factor for CVD. We found that marijuana smoking activated inflammatory cytokines implicated in CVD. In silico virtual screening identified genistein, a soybean isoflavone, as a putative CB1 antagonist. Human-induced pluripotent stem cell-derived endothelial cells were used to model Δ9-THC-induced inflammation and oxidative stress via NF-κB signaling. Knockdown of the CB1 receptor with siRNA, CRISPR interference, and genistein attenuated the effects of Δ9-THC. In mice, genistein blocked Δ9-THC-induced endothelial dysfunction in wire myograph, reduced atherosclerotic plaque, and had minimal penetration of the central nervous system. Genistein is a CB1 antagonist that attenuates Δ9-THC-induced atherosclerosis.


Subject(s)
Cannabis , Cardiovascular Diseases , Hallucinogens , Analgesics , Animals , Cannabinoid Receptor Agonists/pharmacology , Dronabinol/pharmacology , Endothelial Cells , Genistein/pharmacology , Genistein/therapeutic use , Inflammation/drug therapy , Mice , Receptor, Cannabinoid, CB1 , Receptors, Cannabinoid
8.
Circ Cardiovasc Imaging ; 14(12): 1122-1132, 2021 12.
Article in English | MEDLINE | ID: mdl-34915729

ABSTRACT

BACKGROUND: Computed tomography-based evaluation of aortic stenosis (AS) by calcium scoring does not consider interleaflet differences in leaflet characteristics. Here, we sought to examine the functional implications of these differences. METHODS: We retrospectively reviewed the computed tomography angiograms of 200 male patients with degenerative calcific AS undergoing transcatheter aortic valve replacement and 20 male patients with normal aortic valves. We compared the computed tomography angiography (CTA)-derived aortic valve leaflet calcification load (AVLCCTA), appearance, and systolic leaflet excursion (LEsys) of individual leaflets. We performed computer simulations of normal valves to investigate how interleaflet differences in LEsys affect aortic valve area. We used linear regression to identify predictors of leaflet-specific calcification in patients with AS. RESULTS: In patients with AS, the noncoronary cusp (NCC) carried the greatest AVLCCTA (365.9 [237.3-595.4] Agatston unit), compared to the left coronary cusp (LCC, 278.5 [169.2-478.8] Agatston unit) and the right coronary cusp (RCC, 240.6 [137.3-439.0] Agatston unit; both P<0.001). However, LCC conferred the least LEsys (42.8° [38.8°-49.0°]) compared to NCC (44.8° [41.1°-49.78°], P=0.001) and RCC (47.7° [42.0°-52.3°], P<0.001) and was more often characterized as predominantly thickened (23.5%) compared to NCC (12.5%) and RCC (16.5%). Computer simulations of normal valves revealed greater reductions in aortic valve area following closures of NCC (-32.2 [-38.4 to -25.8]%) and RCC (-35.7 [-40.2 to -32.9]%) than LCC (-24.5 [-28.5 to -18.3]%; both P<0.001). By linear regression, the AVLCCTA of NCC and RCC, but not LCC, predicted LEsys (both P<0.001) in patients with AS. Both ostial occlusion and ostial height of the right coronary artery predicted AVLCCTA, RCC (P=0.005 and P=0.001). CONCLUSIONS: In male patients, the AVLCCTA of NCC and RCC contribute more to AS than that of LCC. LCC's propensity for noncalcific leaflet thickening and worse LEsys, however, should not be underestimated when using calcium scores to assess AS severity.


Subject(s)
Aortic Valve Stenosis/complications , Aortic Valve Stenosis/diagnostic imaging , Aortic Valve/pathology , Calcinosis/complications , Calcinosis/diagnostic imaging , Computed Tomography Angiography/methods , Preoperative Care/methods , Aged , Aged, 80 and over , Aortic Valve/diagnostic imaging , Cross-Sectional Studies , Humans , Male , Middle Aged , Retrospective Studies
9.
JACC Case Rep ; 3(9): 1177-1181, 2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34401754

ABSTRACT

Coronary artery vasospasm is typically managed through avoidance of triggers and with symptomatic treatments with calcium channel blockers and long-acting nitrates. Here, we report a rare case of medically refractory coronary artery vasospasm associated with genetic predispositions that initially required cardiac autotransplantation followed paradoxically by nicotine for long-term symptomatic control. (Level of Difficulty: Intermediate.).

10.
STAR Protoc ; 2(2): 100556, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34151292

ABSTRACT

Endothelial cells (ECs) have emerged as key pathogenic players in cardiac disease due to their proximity with cardiomyocytes. Induced pluripotent stem cells (iPSCs) have been employed to generate ECs. However, it may be more clinically relevant to transdifferentiate fibroblasts into ECs directly without introducing pluripotent or virally driven transcription factors. Here, we present a protocol that describes the direct conversion of human cardiac fibroblasts into ECs by leveraging the innate immune system. Our protocol produces bona fide human ECs with 95%-98% purity by first passage. For complete details on the use and execution of this protocol, please refer to Liu et al. (2020) and Sayed et al. (2015).


Subject(s)
Cell Transdifferentiation , Immunity, Innate , Myocytes, Cardiac/cytology , Culture Media , Endothelial Cells/cytology , Fibroblasts/cytology , Humans , Transcription Factors/metabolism
11.
J Vasc Med Surg ; 9(7)2021.
Article in English | MEDLINE | ID: mdl-36276915

ABSTRACT

Cigarette smoking is undoubtedly the single most important risk factor and trigger for vasospastic angina, a condition also known as Prinzmetal angina secondary to coronary artery vasospasm. Even decades before vasospastic angina was first described by Dr. Myron Prinzmetal and his colleagues in 1959, there had been suspected connections between smoking and coronary artery vasospasm in what was alluded to then as "tobacco angina." The intimate relationship between smoking and vasospastic angina has since been extensively researched and validated through decades of epidemiological and clinical studies. The fact that smoking would aggravate vasospastic angina comes with very little surprise, as it has been shown to adversely impact many of the disease processes thought to underlie vasospastic angina, including autonomic dysfunction, endothelial dysfunction, smooth muscle hyperactivity, and genetic susceptibility. While avoidance of smoking is the first logical step in managing smokers with vasospastic angina, there have been reported cases of vasospastic angina paradoxically triggered by smoking cessation or relieved with smoking resumption or nicotine replacement therapy. Thus, there appears to be patient-specific factors that could significantly alter the close connection between smoking and vasospastic angina, warranting further mechanistic investigations. In this review, we will examine this complicated relationship between smoking and vasospastic angina from multiple perspectives (historical, mechanistic, and clinical) and call attention to the "smoking paradox," which, with further elucidation, may provide additional insight into the complex mechanisms of VSA and potentially new strategies to treat medically refractory VSA, at least in selected individuals.

12.
Stem Cells ; 38(6): 808-814, 2020 06.
Article in English | MEDLINE | ID: mdl-32129537

ABSTRACT

Little is known on the phenotypic characteristics of stem cells (SCs) after they are transplanted to the myocardium, in part due to lack of noninvasive platforms to study SCs directly in the living subject. Reporter gene imaging has played a valuable role in the noninvasive assessment of cell fate in vivo. In this study, we validated a pathway-specific reporter gene that can be used to noninvasively image the phenotype of SCs transplanted to the myocardium. Rat mesenchymal SCs (MSCs) were studied for phenotypic evidence of myogenic characteristics under in vitro conditions. After markers of myogenic characteristics were identified, we constructed a reporter gene sensor, comprising the firefly luciferase (Fluc) reporter gene driven by the troponin T (TnT) promoter (cardio MSCs had threefold expression in polymerase chain reaction compared to control MSCs) using a two-step signal amplification strategy. MSCs transfected with TnT-Fluc were studied and validated under in vitro conditions, showing a strong signal after MSCs acquired myogenic characteristics. Lastly, we observed that cardio MSCs had higher expression of the reporter sensor compared to control cells (0.005 ± 0.0005 vs 0.0025 ± 0.0008 Tnt-Fluc/ubiquitin-Fluc, P < .05), and that this novel sensor can detect the change in the phenotype of MSCs directly in the living subject. Pathway-specific reporter gene imaging allows assessment of changes in the phenotype of MSCs after delivery to the ischemic myocardium, providing important information on the phenotype of these cells. Imaging sensors like the one described here are critical to better understanding of the changes that SCs undergo after transplantation.


Subject(s)
Genes, Reporter/genetics , Mesenchymal Stem Cells/metabolism , Cell Differentiation , Humans
13.
Stem Cell Reports ; 14(2): 192-200, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32048999

ABSTRACT

Innate immune signaling has recently been shown to play an important role in nuclear reprogramming, by altering the epigenetic landscape and thereby facilitating transcription. However, the mechanisms that link innate immune activation and metabolic regulation in pluripotent stem cells remain poorly defined, particularly with regard to key molecular components. In this study, we show that hypoxia-inducible factor 1α (HIF1α), a central regulator of adaptation to limiting oxygen tension, is an unexpected but crucial regulator of innate immune-mediated nuclear reprogramming. HIF1α is dramatically upregulated as a consequence of Toll-like receptor 3 (TLR3) signaling and is necessary for efficient induction of pluripotency and transdifferentiation. Bioenergetics studies reveal that HIF1α regulates the reconfiguration of innate immune-mediated reprogramming through its well-established role in throwing a glycolytic switch. We believe that results from these studies can help us better understand the influence of immune signaling in tissue regeneration and lead to new therapeutic strategies.


Subject(s)
Cell Nucleus/metabolism , Cellular Reprogramming , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity, Innate , Animals , Cell Nucleus/drug effects , Cell Transdifferentiation/drug effects , Cellular Reprogramming/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Immunity, Innate/drug effects , Male , Mice, Knockout , Poly I-C/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism , Transcription, Genetic/drug effects
14.
J Vasc Res ; 56(1): 11-15, 2019.
Article in English | MEDLINE | ID: mdl-30763932

ABSTRACT

Peripartum cardiomyopathy (PPCM) is a rare form of congestive heart failure characterized by left ventricular dysfunction that develops towards the end of pregnancy or during the early postpartum phase. Even though the majority of PPCM patients show partial or complete recovery of their heart functions, the mortality rate of PPCM remains high. Previous research has suggested that vascular dysfunction triggered by late-gestational hormones and potent anti-angiogenic factors play key roles in the pathogenesis of PPCM; however, the exact mechanisms remain elusive due to limited patient tissues for characterization. Here, we report a case of PPCM where the coronary vessels from the patient's explanted heart showed marked vascular dysfunction with impaired nitric oxide response. Importantly, these vessels exhibited deficient adenosine-mediated vasorelaxation when subjected to myograph studies, suggesting impaired Kv7 ion channels. Results from this work may lead to new therapeutic strategies for improving Kv7 function in PPCM patients.


Subject(s)
Cardiomyopathies/etiology , Coronary Artery Disease/etiology , Coronary Vessels/physiopathology , Peripartum Period , Vasodilation , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/physiopathology , Cardiomyopathies/surgery , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/physiopathology , Coronary Artery Disease/surgery , Coronary Vessels/diagnostic imaging , Coronary Vessels/metabolism , Disease Progression , Female , Heart Failure/etiology , Heart Failure/physiopathology , Heart Failure/surgery , Heart Transplantation , Humans , KCNQ Potassium Channels/metabolism , Middle Aged , Pregnancy , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/surgery
15.
Circ Res ; 123(4): 443-450, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29986945

ABSTRACT

RATIONALE: Human-induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) have risen as a useful tool in cardiovascular research, offering a wide gamut of translational and clinical applications. However, inefficiency of the currently available iPSC-EC differentiation protocol and underlying heterogeneity of derived iPSC-ECs remain as major limitations of iPSC-EC technology. OBJECTIVE: Here, we performed droplet-based single-cell RNA sequencing (scRNA-seq) of the human iPSCs after iPSC-EC differentiation. Droplet-based scRNA-seq enables analysis of thousands of cells in parallel, allowing comprehensive analysis of transcriptional heterogeneity. METHODS AND RESULTS: Bona fide iPSC-EC cluster was identified by scRNA-seq, which expressed high levels of endothelial-specific genes. iPSC-ECs, sorted by CD144 antibody-conjugated magnetic sorting, exhibited standard endothelial morphology and function including tube formation, response to inflammatory signals, and production of NO. Nonendothelial cell populations resulting from the differentiation protocol were identified, which included immature cardiomyocytes, hepatic-like cells, and vascular smooth muscle cells. Furthermore, scRNA-seq analysis of purified iPSC-ECs revealed transcriptional heterogeneity with 4 major subpopulations, marked by robust enrichment of CLDN5, APLNR, GJA5, and ESM1 genes, respectively. CONCLUSIONS: Massively parallel, droplet-based scRNA-seq allowed meticulous analysis of thousands of human iPSCs subjected to iPSC-EC differentiation. Results showed inefficiency of the differentiation technique, which can be improved with further studies based on identification of molecular signatures that inhibit expansion of nonendothelial cell types. Subtypes of bona fide human iPSC-ECs were also identified, allowing us to sort for iPSC-ECs with specific biological function and identity.


Subject(s)
Endothelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Transcriptome , Apelin Receptors/genetics , Apelin Receptors/metabolism , Cell Differentiation , Cells, Cultured , Claudin-5/genetics , Claudin-5/metabolism , Connexins/genetics , Connexins/metabolism , Endothelial Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Single-Cell Analysis , Gap Junction alpha-5 Protein
16.
Am J Physiol Lung Cell Mol Physiol ; 313(2): L252-L266, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28473326

ABSTRACT

Pulmonary arterial hypertension is a complication of methamphetamine use (METH-PAH), but the pathogenic mechanisms are unknown. Given that cytochrome P450 2D6 (CYP2D6) and carboxylesterase 1 (CES1) are involved in metabolism of METH and other amphetamine-like compounds, we postulated that loss of function variants could contribute to METH-PAH. Although no difference in CYP2D6 expression was seen by lung immunofluorescence, CES1 expression was significantly reduced in endothelium of METH-PAH microvessels. Mass spectrometry analysis showed that healthy pulmonary microvascular endothelial cells (PMVECs) have the capacity to both internalize and metabolize METH. Furthermore, whole exome sequencing data from 18 METH-PAH patients revealed that 94.4% of METH-PAH patients were heterozygous carriers of a single nucleotide variant (SNV; rs115629050) predicted to reduce CES1 activity. PMVECs transfected with this CES1 variant demonstrated significantly higher rates of METH-induced apoptosis. METH exposure results in increased formation of reactive oxygen species (ROS) and a compensatory autophagy response. Compared with healthy cells, CES1-deficient PMVECs lack a robust autophagy response despite higher ROS, which correlates with increased apoptosis. We propose that reduced CES1 expression/activity could promote development of METH-PAH by increasing PMVEC apoptosis and small vessel loss.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Endothelial Cells/metabolism , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Lung/metabolism , Methamphetamine/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Endothelial Cells/drug effects , Female , Humans , Lung/drug effects , Male , Microvessels/drug effects , Microvessels/metabolism , Middle Aged , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
18.
Circ Res ; 119(1): 16-20, 2016 06 24.
Article in English | MEDLINE | ID: mdl-27340267
19.
Nat Rev Cardiol ; 13(6): 333-49, 2016 06.
Article in English | MEDLINE | ID: mdl-27009425

ABSTRACT

The advent of human induced pluripotent stem cell (hiPSC) technology has revitalized the efforts in the past decade to realize more fully the potential of human embryonic stem cells for scientific research. Adding to the possibility of generating an unlimited amount of any cell type of interest, hiPSC technology now enables the derivation of cells with patient-specific phenotypes. Given the introduction and implementation of the large-scale Precision Medicine Initiative, hiPSC technology will undoubtedly have a vital role in the advancement of cardiovascular research and medicine. In this Review, we summarize the progress that has been made in the field of hiPSC technology, with particular emphasis on cardiovascular disease modelling and drug development. The growing roles of hiPSC technology in the practice of precision medicine will also be discussed.


Subject(s)
Cardiovascular Diseases/drug therapy , Induced Pluripotent Stem Cells , Models, Cardiovascular , Precision Medicine , Cardiovascular Diseases/genetics , Cardiovascular Diseases/prevention & control , Cell Differentiation , Cellular Reprogramming Techniques , Drug Discovery , Endothelial Cells/cytology , Humans , Myocytes, Cardiac/cytology , Myocytes, Smooth Muscle/cytology
20.
EMBO Mol Med ; 7(9): 1090-103, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26183451

ABSTRACT

Heart disease remains a leading cause of mortality and a major worldwide healthcare burden. Recent advances in stem cell biology have made it feasible to derive large quantities of cardiomyocytes for disease modeling, drug development, and regenerative medicine. The discoveries of reprogramming and transdifferentiation as novel biological processes have significantly contributed to this paradigm. This review surveys the means by which reprogramming and transdifferentiation can be employed to generate induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and induced cardiomyocytes (iCMs). The application of these patient-specific cardiomyocytes for both in vitro disease modeling and in vivo therapies for various cardiovascular diseases will also be discussed. We propose that, with additional refinement, human disease-specific cardiomyocytes will allow us to significantly advance the understanding of cardiovascular disease mechanisms and accelerate the development of novel therapeutic options.


Subject(s)
Cardiovascular Diseases/therapy , Cell Transdifferentiation , Myocytes, Cardiac/physiology , Regenerative Medicine/methods , Regenerative Medicine/trends , Stem Cells/physiology , Animals , Biomedical Research/methods , Biomedical Research/trends , Disease Models, Animal , Humans
SELECTION OF CITATIONS
SEARCH DETAIL