Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
J Transl Med ; 22(1): 783, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175068

ABSTRACT

A novel approach to treating heart failures was developed with the introduction of iPSC technology. Knowledge in regenerative medicine, developmental biology, and the identification of illnesses at the cellular level has exploded since the discovery of iPSCs. One of the most frequent causes of mortality associated with cardiovascular disease is the loss of cardiomyocytes (CMs), followed by heart failure. A possible treatment for heart failure involves restoring cardiac function and replacing damaged tissue with healthy, regenerated CMs. Significant strides in stem cell biology during the last ten years have transformed the in vitro study of human illness and enhanced our knowledge of the molecular pathways underlying human disease, regenerative medicine, and drug development. We seek to examine iPSC advancements in disease modeling, drug discovery, iPSC-Based cell treatments, and purification methods in this article.


Subject(s)
Induced Pluripotent Stem Cells , Regeneration , Humans , Induced Pluripotent Stem Cells/cytology , Animals , Heart/physiology , Myocytes, Cardiac/cytology , Regenerative Medicine/methods
2.
Sci Rep ; 14(1): 18063, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117679

ABSTRACT

In recent years, research on organ-on-a-chip technology has been flourishing, particularly for drug screening and disease model development. Fibroblasts and vascular endothelial cells engage in crosstalk through paracrine signaling and direct cell-cell contact, which is essential for the normal development and function of the heart. Therefore, to faithfully recapitulate cardiac function, it is imperative to incorporate fibroblasts and vascular endothelial cells into a heart-on-a-chip model. Here, we report the development of a human heart-on-a-chip composed of induced pluripotent stem cell (iPSC)-derived cardiomyocytes, fibroblasts, and vascular endothelial cells. Vascular endothelial cells cultured on microfluidic channels responded to the flow of culture medium mimicking blood flow by orienting themselves parallel to the flow direction, akin to in vivo vascular alignment in response to blood flow. Furthermore, the flow of culture medium promoted integrity among vascular endothelial cells, as evidenced by CD31 staining and lower apparent permeability. The tri-culture condition of iPSC-derived cardiomyocytes, fibroblasts, and vascular endothelial cells resulted in higher expression of the ventricular cardiomyocyte marker IRX4 and increased contractility compared to the bi-culture condition with iPSC-derived cardiomyocytes and fibroblasts alone. Such tri-culture-derived cardiac tissues exhibited cardiac responses similar to in vivo hearts, including an increase in heart rate upon noradrenaline administration. In summary, we have achieved the development of a heart-on-a-chip composed of cardiomyocytes, fibroblasts, and vascular endothelial cells that mimics in vivo cardiac behavior.


Subject(s)
Endothelial Cells , Fibroblasts , Induced Pluripotent Stem Cells , Lab-On-A-Chip Devices , Myocytes, Cardiac , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Cell Differentiation , Cells, Cultured , Coculture Techniques/methods , Microphysiological Systems
3.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39126016

ABSTRACT

A thorough characterization of induced pluripotent stem cells (iPSCs) used with in vitro models or therapeutics is essential. Even iPSCs derived from a single donor can exhibit variability within and between cell lines, which can lead to heterogeneity in results and hinder the promising future of cell replacement therapies. In this study, the cell seeding density of human and rhesus monkey iPSCs was tested to maximize the cell line-specific yield of the generated cardiomyocytes. We found that, despite using the same iPSC generation and differentiation protocols, the cell seeding density for the cell line-specific best differentiation efficiency could differ by a factor of four for the four cell lines used here. In addition, the cell lines showed differences in the range of cell seeding densities that they could tolerate without the severe loss of differentiation efficiency. Overall, our data show that the cell seeding density is a critical parameter for the differentiation inefficiency of primate iPSCs to cardiomyocytes and that iPSCs generated with the same episomal approach still exhibit considerable heterogeneity. Therefore, individual characterization of iPSC lines is required, and functional comparability with in vivo processes must be ensured to warrant the translatability of in vitro research with iPSCs.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Macaca mulatta , Myocytes, Cardiac , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Humans , Animals , Cell Line , Cell Count , Cell Culture Techniques/methods , Cells, Cultured
4.
Methods Mol Biol ; 2835: 69-82, 2024.
Article in English | MEDLINE | ID: mdl-39105907

ABSTRACT

Cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) offer an attractive platform to evaluate the mechanisms of cardiovascular-related incidents and to develop and test new drugs for heart diseases. This work focuses on the comparison of two hiPSC-CM differentiation protocols: the GiWi method based on temporal modulation of the Wnt/ß-catenin pathway and the commercially available PSC Cardiomyocyte Differentiation Kit. We underlined the need to optimize several parameters such as cell density or small molecule concentration (CHIR-99021, IWR-1) to obtain functional hiPSC-CMs. Both protocols yield a similar differentiation efficiency; therefore, the choice of a particular procedure may depend on the preferences of the experimenter.


Subject(s)
Cell Culture Techniques , Cell Differentiation , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Cell Culture Techniques/methods , Wnt Signaling Pathway , Cells, Cultured
5.
Methods Mol Biol ; 2835: 83-98, 2024.
Article in English | MEDLINE | ID: mdl-39105908

ABSTRACT

Cardiomyocytes (CMs) derived from human-induced pluripotent stem cells (hiPSCs) are considered a promising platform for multiple applications, including disease modeling, regenerative medicine, screening of drug toxicity and investigation of cardiomyogenesis. Despite remarkable improvement in methodology enabling differentiation of hiPSCs into CMs, applied protocols generate heterogeneous cell populations composed of CMs along with differentiated non-cardiac cell-types and undifferentiated hiPSCs. Here we describea procedure of automated Magnetic-Activated Cell Sorting (autoMACS) for the purification of hiPSCs-derived CMs under sterile culture conditions. We illustrate that this approach led to a robust depletion of non-cardiac cells and enrichment of CMs, a result particularly crucial for hiPSC lines with poor cardiac differentiation efficiencies.


Subject(s)
Cell Differentiation , Flow Cytometry , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Flow Cytometry/methods , Cell Separation/methods , Cell Culture Techniques/methods , Immunomagnetic Separation/methods , Cells, Cultured
6.
Methods Mol Biol ; 2835: 147-154, 2024.
Article in English | MEDLINE | ID: mdl-39105913

ABSTRACT

Metabolism has emerged recently as an important determinant of stem cell function. Changes in metabolic signaling pathways precede changes in stem cell molecular and functional response. Pluripotent stem cells are highly proliferative and known to exhibit increased glycolysis. Similarly, adult stem cells reside in tissue niches in a quiescent state operating via glycolysis. Upon activation, adult stem cell metabolism transitions from glycolysis to oxidative phosphorylation which coincides with reduced proliferation and multilineage potential. In the heart, different populations of cardiac progenitor cells (CPCs) have been identified. CPCs regenerative potential is linked to changes in metabolic characteristics of cells, impacting cardiac repair following injury. Here, we discuss the methodologies for isolation and characterization of a novel cardiac progenitor cell population from the heart including measurement its metabolic features.


Subject(s)
Stem Cells , Animals , Stem Cells/metabolism , Stem Cells/cytology , Glycolysis , Myocardium/metabolism , Myocardium/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Mice , Humans , Cell Differentiation , Oxidative Phosphorylation , Cell Proliferation , Cell Separation/methods
7.
Methods Mol Biol ; 2835: 121-133, 2024.
Article in English | MEDLINE | ID: mdl-39105911

ABSTRACT

Leigh syndrome (LS), a complex multisystemic disorder, poses significant challenges in genetic medicine due to its intricate pathogenesis and wide-ranging clinical manifestations. Notably, these arise from mutations in either nuclear genetic DNA or mitochondrial DNA, affecting ATP production and resulting in diverse clinical outcomes. The unpredictable trajectory of this disease, ranging from severe developmental delays to early mortality, underscores the need for improved therapeutic solutions. This research pivots toward the novel use of induced pluripotent stem cells (iPSCs) as a promising platform for understanding disease mechanisms and spearheading patient-specific drug discoveries. Given the past successes of iPSCs in delineating organ-specific disorders and the recent endorsement of human iPSC-derived cardiomyocytes (CMs) by the FDA for drug evaluation, our work seeks to bridge this innovation to Leigh syndrome research. We detail a methodological approach to generate iPSCs from LS patients and differentiate them into iPSCs-CMs. Using multi-electrode array (MEA) analyses, we evaluate the field potential of these cells, spotlighting the potential of hiPSC-CM in drug validation and disease modeling. This pioneering approach offers a glimpse into the future of patient-centric therapeutic interventions for Leigh/Leigh-like syndrome.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Leigh Disease , Myocytes, Cardiac , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Humans , Leigh Disease/genetics , Leigh Disease/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Cells, Cultured , Cell Culture Techniques/methods , Drug Evaluation, Preclinical/methods
8.
Methods Mol Biol ; 2835: 269-275, 2024.
Article in English | MEDLINE | ID: mdl-39105922

ABSTRACT

Three-dimensional (3D) scaffolds provide cell support while improving tissue regeneration through amplified cellular responses between implanted materials and native tissues. So far, highly conductive cardiac, nerve, and muscle tissues have been engineered by culturing stem cells on electrically inert scaffolds. These scaffolds, even though suitable, may not be very useful compared to the results shown by cells when cultured on conductive scaffolds. Noticing the mature phenotype the stem cells develop over time when cultured on conductive scaffolds, scientists have been trying to impart conductivity to traditionally nonconductive scaffolds. One way to achieve this goal is to blend conductive polymers (polyaniline, polypyrrole, PEDOT:PSS) with inert biomaterials and produce a 3D scaffold using various fabrication techniques. One such technique is projection micro-stereolithography, which is an additive manufacturing technique. It uses a photosensitive solution blended with conductive polymers and uses visible/UV light to crosslink the solution. 3D scaffolds with complex architectural features down to microscale resolution can be printed with this technique promptly. This chapter reports a protocol to fabricate electrically conductive scaffolds using projection micro-stereolithography.


Subject(s)
Cell Culture Techniques , Electric Conductivity , Polymers , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Polymers/chemistry , Cell Culture Techniques/methods , Pyrroles/chemistry , Animals , Humans , Biocompatible Materials/chemistry , Cells, Cultured , Stem Cells/cytology , Aniline Compounds/chemistry , Myocytes, Cardiac/cytology , Stereolithography
9.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(3): 630-634, 2024 May 20.
Article in Chinese | MEDLINE | ID: mdl-38948270

ABSTRACT

Objective: To investigate the effect of Sanshentongmai (SSTM) mixture on the regulation of oxidative damage to rat cardiomyocytes (H9C2) through microRNA-146a and its mechanism. Methods: H9C2 were cultured in vitro, H2O2 was used as an oxidant to create an oxidative damage model in H9C2 cells. SSTM intervention was administered to the H9C2 cells. Then, the changes in H2O2-induced oxidative damage in H9C2 cells and the expression of microRNA-146a were observed to explore the protective effect of SSTM on H9C2 and its mechanism. H9C2 cells cultured i n vitro were divided into 3 groups, including a control group, a model group of H2O2-induced oxidative damage (referred to hereafter as the model group), and a group given H2O2 modeling plus SSTM intervention at 500 µg/L for 72 h (referred to hereafter as the treatment group). The cell viability was measured by CCK8 assay. In addition, the levels of N-terminal pro-brain natriuretic peptide (Nt-proBNP), nitric oxide (NO), high-sensitivity C-reactive protein (Hs-CRP), and angiotensin were determined by enzyme-linked immunosorbent assay (ELISA). The expression level of microRNA-146a was determined by real-time PCR (RT-PCR). Result: H9C2 cells were pretreated with SSTM at mass concentrations ranging from 200 to 1500 µg/L. Then, CCK8 assay was performed to measure cell viability and the findings showed that the improvement in cell proliferation reached its peak when the mass concentration of SSTM was 500 µg/L, which was subsequently used as the intervention concentration. ELISA was performed to measure the indicators related to heart failure, including Nt-proBNP, NO, Hs-CRP, and angiotensin Ⅱ. Compared with those of the control group, the expressions of Nt-proBNP and angiotensin Ⅱ in the treatment group were up-regulated (P<0.05), while the expression of NO was down-regulated (P<0.05). There was no significant difference in the expression of Hs-CRP between the treatment group and the control group. These findings indicate that SSTM could effectively ameliorate oxidative damage in H9C2 rat cardiomyocytes. Finally, according to the RT-PCR findings for the expression of microRNA-146a in each group, H2O2 treatment at 15 µmol/L could significantly reduce the expression of microRNA-146a, and the expression of microRNA-146a in the treatment group was nearly doubled compared with that in the model group. There was no significant difference between the treatment group and the control group. Conclusion: SSTM can significantly resist the H2O2-induced oxidative damage of H9C2 cells and may play a myocardial protective role by upregulating microRNA-146a.


Subject(s)
Drugs, Chinese Herbal , Hydrogen Peroxide , MicroRNAs , Myocytes, Cardiac , Oxidative Stress , Up-Regulation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/cytology , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Rats , Oxidative Stress/drug effects , Hydrogen Peroxide/toxicity , Drugs, Chinese Herbal/pharmacology , Up-Regulation/drug effects , Cell Survival/drug effects , Cell Line , Drug Combinations
10.
Cell Biochem Funct ; 42(5): e4090, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38973147

ABSTRACT

Cellular therapy is considered a better option for the treatment of degenerative disorders. Different cell types are being used for tissue regeneration. Despite extensive research in this field, several issues remain to be addressed concerning cell transplantation. One of these issues is the survival and homing of administered cells in the injured tissue, which depends on the ability of these cells to adhere. To enhance cell adherence and survival, Rap1 GTPase was activated in mesenchymal stem cells (MSCs) as well as in cardiomyocytes (CMs) by using 8-pCPT-2'-O-Me-cAMP, and the effect on gene expression dynamics was determined through quantitative reverse transcriptase-polymerase chain reaction analysis. Pharmacological activation of MSCs and CMs resulted in the upregulation of connexin-43 and cell adhesion genes, which increased the cell adhesion ability of MSCs and CMs, and increased the fusion of MSCs with neonatal CMs. Treating stem cells with a pharmacological agent that activates Rap1a before transplantation can enhance their fusion with CMs and increase cellular regeneration.


Subject(s)
Mesenchymal Stem Cells , Myocytes, Cardiac , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Animals , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Cell Fusion , Cells, Cultured , Rats , Animals, Newborn , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/genetics
11.
Stem Cell Res Ther ; 15(1): 213, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39020441

ABSTRACT

BACKGROUND: Commonly used media for the differentiation of human pluripotent stem cells into cardiomyocytes (hPSC-CMs) contain high concentrations of proteins, in particular albumin, which is prone to quality variations and presents a substantial cost factor, hampering the clinical translation of in vitro-generated cardiomyocytes for heart repair. To overcome these limitations, we have developed chemically defined, entirely protein-free media based on RPMI, supplemented with L-ascorbic acid 2-phosphate (AA-2P) and either the non-ionic surfactant Pluronic F-68 or a specific polyvinyl alcohol (PVA). METHODS AND RESULTS: Both media compositions enable the efficient, directed differentiation of embryonic and induced hPSCs, matching the cell yields and cardiomyocyte purity ranging from 85 to 99% achieved with the widely used protein-based CDM3 medium. The protein-free differentiation approach was readily up-scaled to a 2000 mL process scale in a fully controlled stirred tank bioreactor in suspension culture, producing > 1.3 × 109 cardiomyocytes in a single process run. Transcriptome analysis, flow cytometry, electrophysiology, and contractile force measurements revealed that the mass-produced cardiomyocytes differentiated in protein-free medium exhibit the expected ventricular-like properties equivalent to the well-established characteristics of CDM3-control cells. CONCLUSIONS: This study promotes the robustness and upscaling of the cardiomyogenic differentiation process, substantially reduces media costs, and provides an important step toward the clinical translation of hPSC-CMs for heart regeneration.


Subject(s)
Cell Differentiation , Culture Media , Myocytes, Cardiac , Humans , Cell Differentiation/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Cell Culture Techniques/methods , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/drug effects , Cells, Cultured
12.
Cell Stem Cell ; 31(7): 941-943, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38971143

ABSTRACT

Induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) therapy has emerged as a highly promising field of heart repair. Lin et al.1 presented compelling evidence on the long-term engraftment and maturation of autologous iPSC-CMs in two rhesus macaques, demonstrating unprecedented cardiac autografting data in large animal models without the need of immunosuppressants.


Subject(s)
Induced Pluripotent Stem Cells , Macaca mulatta , Myocytes, Cardiac , Animals , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Autografts , Humans , Cell Survival , Cell Differentiation
13.
Methods Mol Biol ; 2805: 89-100, 2024.
Article in English | MEDLINE | ID: mdl-39008175

ABSTRACT

Engineered heart tissues (EHTs) have been shown to be a valuable platform for disease investigation and therapeutic testing by increasing human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) maturity and better recreating the native cardiac environment. The protocol detailed in this chapter describes the generation of miniaturized EHTs (mEHTs) incorporating hiPSC-CMs and human stromal cells in a fibrin hydrogel. This platform utilizes an array of silicone posts designed to fit in a standard 96-well tissue culture plate. Stromal cells and hiPSC-CMs are cast in a fibrin matrix suspended between two silicone posts, forming an mEHT that produces synchronous muscle contractions. The platform presented here has the potential to be used for high throughput characterization and screening of disease phenotypes and novel therapeutics through measurements of the myocardial function, including contractile force and calcium handling, and its compatibility with immunostaining.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Tissue Engineering , Humans , Tissue Engineering/methods , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Hydrogels/chemistry , Cell Differentiation , Fibrin/metabolism , Cells, Cultured , Cell Culture Techniques/methods , Stromal Cells/cytology , Tissue Culture Techniques/methods , Tissue Culture Techniques/instrumentation
14.
Sci Rep ; 14(1): 16966, 2024 07 23.
Article in English | MEDLINE | ID: mdl-39043870

ABSTRACT

Stem cells' differentiation toward cardiac lineage is a complex process dependent on various alterations in molecular basis and regulation pathways. The aim of the study is to show that endometrium-derived stromal cells - menstrual, endometrial and endometriotic, could be an attractive source for examination of the mechanisms underlying cardiomyogenesis. After treatment with Decitabine, Angiotensin II and TGF-ß1, cells demonstrated morphological dedifferentiation into early cardiomyocyte-like cells and expressed CD36, CD106, CD172a typically used to sort for human pluripotent stem cell-derived cardiomyocytes. RT-qPCR revealed changed cells' genetic profiles, as majority of cardiac lineage differentiation related genes and cardiac ion channels (calcium, sodium, potassium) coding genes were upregulated after 6 and 13 days of exposure. Additionally, analysis of expression of various signaling proteins (FOXO1, PDGFB, TGFBR1, mTOR, VEGFA, WNT4, Notch1) coding genes showed differences between cell cultures as they seem to employ distinct signaling pathways through differentiation initiation. Early stages of differentiation had biggest impact on cardiomyogenesis related proteins (Nkx-2.5, EZH2, FOXO3a, H3K9Ac) levels, as we noticed after conducting Western blot and as expected, early cardiac transcription factor Nkx-2.5 was highly expressed and localized in nucleus of differentiating cells. These findings led us to assess endometrium origin stromal cells' potential to differentiate towards cardiomyogenic lineage and better understand the regulation of complex differentiation processes in ex vivo model systems.


Subject(s)
Angiotensin II , Cell Differentiation , Decitabine , Endometrium , Myocytes, Cardiac , Stromal Cells , Transforming Growth Factor beta1 , Humans , Female , Cell Differentiation/drug effects , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Endometrium/cytology , Endometrium/metabolism , Endometrium/drug effects , Stromal Cells/metabolism , Stromal Cells/drug effects , Stromal Cells/cytology , Angiotensin II/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Decitabine/pharmacology , Cells, Cultured , Adult , Signal Transduction/drug effects
15.
Tissue Cell ; 89: 102461, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38991272

ABSTRACT

The current study was constructed to fabricate polyamide based nanofibrous scaffolds (NS) and to define the most promising one for the generation of cardiomyocytes from adipose tissue derived mesenchymal stem cells (ADMSCs). This purpose was extended to assess the potentiality of the generated cardiomyocytes in relieving myocardial infarction (MI) in rats. Production and characterization of NSs were carried out. ADMSCs were cultured on NS and induced to differentiate into cardiomyocytes by specific growth factors. Molecular analysis for myocyte-specific enhancer factor 2 C (MEF2C) and alpha sarcomeric actin (α-SCA) expression was done to confirm the differentiation of ADMSCs into cardiomyocytes for further transplantation into MI induced rats. Implantation of cells in MI afflicted rats boosted heart rate, ST height and PR interval and lessened P duration, RR, QTc and QRS intervals. Also, this type of medication minified serum lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) enzymes activity as well as serum and cardiac troponin T (Tn-T) levels and upraised serum and cardiac α-SCA and cardiac connexin 43 (CX 43) levels. Microscopic feature of cardiac tissue sections of rats in the treated groups revealed great renovation in the cardiac microarchitecture. Conclusively, this attempt gains insight into a realistic strategy for recovery of MI through systemic employment of in vitro generated cardiomyocytes.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , Myocardial Infarction , Myocytes, Cardiac , Nanofibers , Tissue Scaffolds , Animals , Myocardial Infarction/therapy , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Tissue Scaffolds/chemistry , Nanofibers/chemistry , Rats , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Cells, Cultured , Male
16.
Int J Mol Sci ; 25(14)2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39063213

ABSTRACT

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have emerged as a promising tool for studying cardiac physiology and drug responses. However, their use is largely limited by an immature phenotype and lack of high-throughput analytical methodology. In this study, we developed a high-throughput testing platform utilizing hPSC-CMs to assess the cardiotoxicity and effectiveness of drugs. Following an optimized differentiation and maturation protocol, hPSC-CMs exhibited mature CM morphology, phenotype, and functionality, making them suitable for drug testing applications. We monitored intracellular calcium dynamics using calcium imaging techniques to measure spontaneous calcium oscillations in hPSC-CMs in the presence or absence of test compounds. For the cardiotoxicity test, hPSC-CMs were treated with various compounds, and calcium flux was measured to evaluate their effects on calcium dynamics. We found that cardiotoxic drugs withdrawn due to adverse drug reactions, including encainide, mibefradil, and cetirizine, exhibited toxicity in hPSC-CMs but not in HEK293-hERG cells. Additionally, in the effectiveness test, hPSC-CMs were exposed to ATX-II, a sodium current inducer for mimicking long QT syndrome type 3, followed by exposure to test compounds. The observed changes in calcium dynamics following drug exposure demonstrated the utility of hPSC-CMs as a versatile model system for assessing both cardiotoxicity and drug efficacy. Overall, our findings highlight the potential of hPSC-CMs in advancing drug discovery and development, which offer a physiologically relevant platform for the preclinical screening of novel therapeutics.


Subject(s)
Cell Differentiation , Drug Evaluation, Preclinical , Myocytes, Cardiac , Pluripotent Stem Cells , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Drug Evaluation, Preclinical/methods , Cell Differentiation/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cardiovascular Agents/pharmacology , Calcium/metabolism , Cardiotoxicity , High-Throughput Screening Assays/methods , HEK293 Cells , Calcium Signaling/drug effects
17.
Curr Protoc ; 4(7): e1097, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39036931

ABSTRACT

In the heart in vivo, vasculature forms a semi-permeable endothelial barrier for selective nutrient and (immune) cell delivery to the myocardium and removal of waste products. Crosstalk between the vasculature and the heart cells regulates homeostasis in health and disease. To model heart development and disease in vitro it is important that essential features of this crosstalk are captured. Cardiac organoid and microtissue models often integrate endothelial cells (ECs) to form microvascular networks inside the 3D structure. However, in static culture without perfusion, these networks may fail to show essential functionality. Here, we describe a protocol to generate an in vitro model of human induced pluripotent stem cell (hiPSC)-derived vascularized cardiac microtissues on a microfluidic organ-on-chip platform (VMToC) in which the blood vessels are perfusable. First, prevascularized cardiac microtissues (MT) are formed by combining hiPSC-derived cardiomyocytes, ECs, and cardiac fibroblasts in a pre-defined ratio. Next, these prevascularized MTs are integrated in the chips in a fibrin hydrogel containing additional vascular cells, which self-organize into tubular structures. The MTs become vascularized through anastomosis between the pre-existing microvasculature in the MT and the external vascular network. The VMToCs are then ready for downstream structural and functional assays and basic characterization. Using this protocol, cardiac MTs can be efficiently and robustly vascularized and perfused within 7 days. In vitro vascularized organoid and MT models have the potential to transition current 3D cardiac models to more physiologically relevant organ models that allow the role of the endothelial barrier in drug and inflammatory response to be investigated. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Generation of VMToC Support Protocol 1: Functional Characterization of VMToC Support Protocol 2: Structural Characterization of VMToC.


Subject(s)
Induced Pluripotent Stem Cells , Lab-On-A-Chip Devices , Myocytes, Cardiac , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Endothelial Cells/cytology , Myocardium/cytology , Myocardium/metabolism , Cell Differentiation
18.
ACS Appl Mater Interfaces ; 16(28): 36030-36046, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38951110

ABSTRACT

Differentiation of induced pluripotent stem cells (iPSCs) is an extremely complex process that has proven difficult to study. In this research, we utilized nanotopography to elucidate details regarding iPSC differentiation by developing a nanodot platform consisting of nanodot arrays of increasing diameter. Subjecting iPSCs cultured on the nanodot platform to a cardiomyocyte (CM) differentiation protocol revealed several significant gene expression profiles that were associated with poor differentiation. The observed expression trends were used to select existing small-molecule drugs capable of modulating differentiation efficiency. BRD K98 was repurposed to inhibit CM differentiation, while iPSCs treated with NSC-663284, carmofur, and KPT-330 all exhibited significant increases in not only CM marker expression but also spontaneous beating, suggesting improved CM differentiation. In addition, quantitative polymerase chain reaction was performed to determine the gene regulation responsible for modulating differentiation efficiency. Multiple genes involved in extracellular matrix remodeling were correlated with a CM differentiation efficiency, while genes involved in the cell cycle exhibited contrasting expression trends that warrant further studies. The results suggest that expression profiles determined via short time-series expression miner analysis of nanodot-cultured iPSC differentiation can not only reveal drugs capable of enhancing differentiation efficiency but also highlight crucial sets of genes related to processes such as extracellular matrix remodeling and the cell cycle that can be targeted for further investigation. Our findings confirm that the nanodot platform can be used to reveal complex mechanisms behind iPSC differentiation and could be an indispensable tool for optimizing iPSC technology for clinical applications.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Humans , Nanoparticles/chemistry , Cells, Cultured , Nanostructures/chemistry
19.
Stem Cell Reports ; 19(8): 1053-1060, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39059379

ABSTRACT

Transplantation of pluripotent stem cell-derived cardiomyocytes is a novel promising cell-based therapeutic approach for patients with heart failure. However, engraftment arrhythmias are a predictable life-threatening complication and represent a major hurdle for clinical translation. Thus, we wanted to experimentally study whether impulse generation by transplanted cardiomyocytes can propagate to the host myocardium and overdrive the recipient rhythm. We transplanted human induced pluripotent stem cell-derived cardiomyocytes expressing the optogenetic actuator Bidirectional Pair of Opsins for Light-induced Excitation and Silencing (BiPOLES) in a guinea pig injury model. Eight weeks after transplantation ex vivo, Langendorff perfusion was used to assess electrical coupling. Pulsed photostimulation was applied to specifically activate the engrafted cardiomyocytes. Photostimulation resulted in ectopic pacemaking that propagated to the host myocardium, caused non-sustained arrhythmia, and stimulated the recipient heart with higher pacing frequency (4/9 hearts). Our study demonstrates that transplanted cardiomyocytes can (1) electrically couple to the host myocardium and (2) stimulate the recipient heart. Thus, our results provide experimental evidence for an important aspect of engraftment-induced arrhythmia induction and thereby support the current hypothesis that cardiomyocyte automaticity can serve as a trigger for ventricular arrhythmias.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Animals , Guinea Pigs , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Humans , Arrhythmias, Cardiac/therapy , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Optogenetics/methods , Heart/physiology , Cell Differentiation , Myocardium/metabolism , Myocardium/cytology , Stem Cell Transplantation/methods
20.
Chem Res Toxicol ; 37(8): 1428-1444, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39046974

ABSTRACT

Environmental chemicals may contribute to the global burden of cardiovascular disease, but experimental data are lacking to determine which substances pose the greatest risk. Human-induced pluripotent stem cell (iPSC)-derived cardiomyocytes are a high-throughput cardiotoxicity model that is widely used to test drugs and chemicals; however, most studies focus on exploring electro-physiological readouts. Gene expression data may provide additional molecular insights to be used for both mechanistic interpretation and dose-response analyses. Therefore, we hypothesized that both transcriptomic and functional data in human iPSC-derived cardiomyocytes may be used as a comprehensive screening tool to identify potential cardiotoxicity hazards and risks of the chemicals. To test this hypothesis, we performed concentration-response analysis of 464 chemicals from 12 classes, including both pharmaceuticals and nonpharmaceutical substances. Functional effects (beat frequency, QT prolongation, and asystole), cytotoxicity, and whole transcriptome response were evaluated. Points of departure were derived from phenotypic and transcriptomic data, and risk characterization was performed. Overall, 244 (53%) substances were active in at least one phenotype; as expected, pharmaceuticals with known cardiac liabilities were the most active. Positive chronotropy was the functional phenotype activated by the largest number of tested chemicals. No chemical class was particularly prone to pose a potential hazard to cardiomyocytes; a varying proportion (10-44%) of substances in each class had effects on cardiomyocytes. Transcriptomic data showed that 69 (15%) substances elicited significant gene expression changes; most perturbed pathways were highly relevant to known key characteristics of human cardiotoxicants. The bioactivity-to-exposure ratios showed that phenotypic- and transcriptomic-based POD led to similar results for risk characterization. Overall, our findings demonstrate how the integrative use of in vitro transcriptomic and phenotypic data from iPSC-derived cardiomyocytes not only offers a complementary approach for hazard and risk prioritization, but also enables mechanistic interpretation of the in vitro test results to increase confidence in decision-making.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Transcriptome , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Transcriptome/drug effects , Environmental Pollutants/toxicity , Dose-Response Relationship, Drug , Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL