Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (418)

Search Parameters:
Keywords = cardiomyocyte differentiation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
39 pages, 1418 KiB  
Review
Human-Induced Pluripotent Stem Cells (iPSCs) for Disease Modeling and Insulin Target Cell Regeneration in the Treatment of Insulin Resistance: A Review
by Sama Thiab, Juberiya M. Azeez, Alekya Anala, Moksha Nanda, Somieya Khan, Alexandra E. Butler and Manjula Nandakumar
Cells 2025, 14(15), 1188; https://doi.org/10.3390/cells14151188 - 1 Aug 2025
Viewed by 122
Abstract
Diabetes mellitus, both type 1 (T1D) and type 2 (T2D), has become the epidemic of the century and a major public health concern given its rising prevalence and the increasing adoption of a sedentary lifestyle globally. This multifaceted disease is characterized by impaired [...] Read more.
Diabetes mellitus, both type 1 (T1D) and type 2 (T2D), has become the epidemic of the century and a major public health concern given its rising prevalence and the increasing adoption of a sedentary lifestyle globally. This multifaceted disease is characterized by impaired pancreatic beta cell function and insulin resistance (IR) in peripheral organs, namely the liver, skeletal muscle, and adipose tissue. Additional insulin target tissues, including cardiomyocytes and neuronal cells, are also affected. The advent of stem cell research has opened new avenues for tackling this disease, particularly through the regeneration of insulin target cells and the establishment of disease models for further investigation. Human-induced pluripotent stem cells (iPSCs) have emerged as a valuable resource for generating specialized cell types, such as hepatocytes, myocytes, adipocytes, cardiomyocytes, and neuronal cells, with diverse applications ranging from drug screening to disease modeling and, importantly, treating IR in T2D. This review aims to elucidate the significant applications of iPSC-derived insulin target cells in studying the pathogenesis of insulin resistance and T2D. Furthermore, recent differentiation strategies, protocols, signaling pathways, growth factors, and advancements in this field of therapeutic research for each specific iPSC-derived cell type are discussed. Full article
(This article belongs to the Special Issue Advances in Human Pluripotent Stem Cells)
Show Figures

Figure 1

18 pages, 1278 KiB  
Review
Metabolic Maturation in hiPSC-Derived Cardiomyocytes: Emerging Strategies for Inducing the Adult Cardiac Phenotype
by Daniela Malan, Maria Pia Gallo, Federica Geddo, Renzo Levi and Giulia Querio
Pharmaceuticals 2025, 18(8), 1133; https://doi.org/10.3390/ph18081133 - 29 Jul 2025
Viewed by 306
Abstract
Human induced pluripotent stem cells (hiPSCs) are widely used in basic research because of their versatility and ability to differentiate into multiple cell types. In particular, differentiating hiPSCs into cardiac cells (hiPSC-CMs) has been an important milestone in cardiac pathophysiology studies. Although hiPSC-CMs [...] Read more.
Human induced pluripotent stem cells (hiPSCs) are widely used in basic research because of their versatility and ability to differentiate into multiple cell types. In particular, differentiating hiPSCs into cardiac cells (hiPSC-CMs) has been an important milestone in cardiac pathophysiology studies. Although hiPSC-CMs offer a model for human cardiomyocytes, they still exhibit characteristics linked to the fetal cardiac cell phenotype. One important feature that prevents hiPSC-CMs from being identified as adult cells relates to their metabolism, which is a key factor in defining a mature phenotype capable of sustaining the workload requirements characteristic of fully differentiated cardiomyocytes. This review aims to present the most relevant strategies in terms of culture medium composition, culture times, and 3D culture methods that have been developed to promote the metabolic maturation of hiPSC-CMs, which are now widely used. Defining a standardized and universally accepted protocol would enable the creation of a cellular model for studies of cardiac pathophysiology from a patient-specific perspective and for drug screening. Full article
(This article belongs to the Special Issue Cell Therapy for Cardiac Disease)
Show Figures

Figure 1

29 pages, 1500 KiB  
Review
Nicotinamide Adenine Dinucleotide Supplementation to Alleviate Heart Failure: A Mitochondrial Dysfunction Perspective
by Fan Yu, Huiying Zhao, Lu Luo and Wei Wu
Nutrients 2025, 17(11), 1855; https://doi.org/10.3390/nu17111855 - 29 May 2025
Cited by 1 | Viewed by 2577
Abstract
Heart failure represents the terminal stage in the development of many cardiovascular diseases, and its pathological mechanisms are closely related to disturbances in energy metabolism and mitochondrial dysfunction in cardiomyocytes. In recent years, nicotinamide adenine dinucleotide (NAD+), a core coenzyme involved [...] Read more.
Heart failure represents the terminal stage in the development of many cardiovascular diseases, and its pathological mechanisms are closely related to disturbances in energy metabolism and mitochondrial dysfunction in cardiomyocytes. In recent years, nicotinamide adenine dinucleotide (NAD+), a core coenzyme involved in cellular energy metabolism and redox homeostasis, has been shown to potentially ameliorate heart failure through the regulation of mitochondrial function. This review systematically investigates four core mechanisms of mitochondrial dysfunction in heart failure: imbalance of mitochondrial dynamics, excessive accumulation of reactive oxygen species (ROS) leading to oxidative stress injury, dysfunction of mitochondrial autophagy, and disturbance of Ca2+ homeostasis. These abnormalities collectively exacerbate the progression of heart failure by disrupting ATP production and inducing apoptosis and myocardial fibrosis. NAD+ has been shown to regulate mitochondrial biosynthesis and antioxidant defences through the activation of the deacetylase family (e.g., silent information regulator 2 homolog 1 (SIRT1) and SIRT3) and to increase mitochondrial autophagy to remove damaged mitochondria, thus restoring energy metabolism and redox balance in cardiomyocytes. In addition, the inhibition of NAD+-degrading enzymes (e.g., poly ADP-ribose polymerase (PARP), cluster of differentiation 38 (CD38), and selective androgen receptor modulators (SARMs)) increases the tissue intracellular NAD+ content, and supplementation with NAD+ precursors (e.g., β-nicotinamide mononucleotide (NMN), nicotinamide riboside, etc.) also significantly elevates myocardial NAD+ levels to ameliorate heart failure. This study provides a theoretical basis for understanding the central role of NAD+ in mitochondrial homeostasis and for the development of targeted therapies for heart failure. Full article
(This article belongs to the Special Issue Nutritional Aspects of Cardiovascular Disease Risk Factors)
Show Figures

Figure 1

25 pages, 1118 KiB  
Review
Induced Pluripotent Stem Cells in Cardiomyopathy: Advancing Disease Modeling, Therapeutic Development, and Regenerative Therapy
by Quan Duy Vo, Kazufumi Nakamura, Yukihiro Saito, Satoshi Akagi, Toru Miyoshi and Shinsuke Yuasa
Int. J. Mol. Sci. 2025, 26(11), 4984; https://doi.org/10.3390/ijms26114984 - 22 May 2025
Viewed by 1128
Abstract
Cardiomyopathies are a heterogeneous group of heart muscle diseases that can lead to heart failure, arrhythmias, and sudden cardiac death. Traditional animal models and in vitro systems have limitations in replicating the complex pathology of human cardiomyopathies. Induced pluripotent stem cells (iPSCs) offer [...] Read more.
Cardiomyopathies are a heterogeneous group of heart muscle diseases that can lead to heart failure, arrhythmias, and sudden cardiac death. Traditional animal models and in vitro systems have limitations in replicating the complex pathology of human cardiomyopathies. Induced pluripotent stem cells (iPSCs) offer a transformative platform by enabling the generation of patient-specific cardiomyocytes, thus opening new avenues for disease modeling, drug discovery, and regenerative therapy. This process involves reprogramming somatic cells into iPSCs and subsequently differentiating them into functional cardiomyocytes, which can be characterized using techniques such as electrophysiology, contractility assays, and gene expression profiling. iPSC-derived cardiomyocyte (iPSC-CM) platforms are also being explored for drug screening and personalized medicine, including high-throughput testing for cardiotoxicity and the identification of patient-tailored therapies. While iPSC-CMs already serve as valuable models for understanding disease mechanisms and screening drugs, ongoing advances in maturation and bioengineering are bringing iPSC-based therapies closer to clinical application. Furthermore, the integration of multi-omics approaches and artificial intelligence (AI) is enhancing the predictive power of iPSC models. iPSC-based technologies are paving the way for a new era of personalized cardiology, with the potential to revolutionize the management of cardiomyopathies through patient-specific insights and regenerative strategies. Full article
(This article belongs to the Special Issue Myocardial Disease: Molecular Pathology and Treatments)
Show Figures

Figure 1

23 pages, 8247 KiB  
Article
The Key Role and Mechanism of Oxidative Stress in Hypertrophic Cardiomyopathy: A Systematic Exploration Based on Multi-Omics Analysis and Experimental Validation
by Sijie Zhang, Tianzhi Li, Shiyi Sun, Yujiao Jiang, Yuxin Sun and Yan Meng
Antioxidants 2025, 14(5), 557; https://doi.org/10.3390/antiox14050557 - 7 May 2025
Viewed by 674
Abstract
Hypertrophic cardiomyopathy (HCM), characterised by abnormal ventricular thickening, involves complex mechanisms including gene mutations, calcium dysregulation, mitochondrial dysfunction, and oxidative stress. Oxidative stress plays a pivotal role in the progression of HCM by mediating cardiomyocyte injury and remodelling. This study systematically analysed HCM [...] Read more.
Hypertrophic cardiomyopathy (HCM), characterised by abnormal ventricular thickening, involves complex mechanisms including gene mutations, calcium dysregulation, mitochondrial dysfunction, and oxidative stress. Oxidative stress plays a pivotal role in the progression of HCM by mediating cardiomyocyte injury and remodelling. This study systematically analysed HCM transcriptomic data using differential gene expression, weighted gene co-expression network analysis (WGCNA), and unsupervised consensus clustering to identify key genes and classify HCM subtypes. Four oxidative stress-related characteristic genes (DUSP1, CCND1, STAT3, and THBS1) were identified using LASSO regression, SVM-RFE, and Random Forest algorithms. Their functional significance was validated by immune infiltration analysis, drug prediction using the cMAP database, and molecular docking. Single-cell RNA sequencing revealed their cell-type-specific expression, and in vitro experiments confirmed their role in HCM. These findings provide insights into oxidative stress mechanisms and potential therapeutic targets for HCM. Full article
Show Figures

Figure 1

18 pages, 6289 KiB  
Article
Regulation of Immune-Related Gene Expression by Salinity-Induced HPI Axis in Large Yellow Croaker, Larimichthys crocea
by Jia Cheng, Zhengjia Lou, Huijie Feng, Yu Zhang, Honghui Li, Wuying Chu and Liangyi Xue
Int. J. Mol. Sci. 2025, 26(9), 4298; https://doi.org/10.3390/ijms26094298 - 1 May 2025
Viewed by 443
Abstract
Large yellow croaker is one of the most popular economic fish species in China. There are studies on the effects of salinity on the growth and development of large yellow croaker (Larimichthys crocea), but the effects of the hypothalamic–pituitary–interrenal axis (HPI), [...] Read more.
Large yellow croaker is one of the most popular economic fish species in China. There are studies on the effects of salinity on the growth and development of large yellow croaker (Larimichthys crocea), but the effects of the hypothalamic–pituitary–interrenal axis (HPI), HPI axis-related genes, and immune-related gene expression and its mechanisms have not been reported. This study analyzed the comparative transcriptomics of brain tissue in large yellow croaker under different salinity (12, 24, and 36 ppt) treatments for 4 weeks. The results showed that there were 1568 differential expression genes in the high salinity (HB) and normal salinity (NB) groups, including 494 up-regulated and 1074 down-regulated transcripts, and 1720 differential expression genes in the low salinity (LB) and normal salinity (NB) groups, including 486 up-regulated and 1234 down-regulated transcripts. Some pathways were significantly enriched, including the adrenergic signaling pathway of cardiomyocytes, oxidative phosphorylation, aldosterone synthesis and secretion, chemokine signaling pathway, and cyclic adenosine monophosphate (cAMP) signaling pathway. Quantitative Real-time polymerase chain reaction (qPCR) analysis further confirmed changes in the expression levels of HPI axis-related genes (β2-ADR, GH, and PRL) and significant changes in the expression levels of immune-related genes (IL6st, IL6, CXCL12, CD40, IFNAR1, SOCS2, SOCS6, and IRF1). In summary, this experiment demonstrates that salinity stress can activate the HPI axis and influence its immune function in large yellow croaker. Furthermore, the expression of immune factors during the immune response is regulated by the upstream genes of the HPI axis. The findings of this study are significant for understanding the physiological and immune responses of large yellow croaker to salinity stress. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

47 pages, 3395 KiB  
Review
Understanding the Mechanisms of Chemotherapy-Related Cardiotoxicity Employing hiPSC-Derived Cardiomyocyte Models for Drug Screening and the Identification of Genetic and Epigenetic Variants
by Abhishikt David Solomon, Swarna Dabral, Raman Gulab Brajesh, Billy W. Day, Matea Juric, Jacek Zielonka, Zeljko J. Bosnjak and Tarun Pant
Int. J. Mol. Sci. 2025, 26(9), 3966; https://doi.org/10.3390/ijms26093966 - 23 Apr 2025
Cited by 1 | Viewed by 2969
Abstract
Chemotherapy-related cardiotoxicity (CTRTOX) is a profound and common side effect of cancer-based therapy in a subset of patients. The underlying factors and the associated mechanisms contributing to severe toxicity of the heart among these patients remain unknown. While challenges remain in accessing human [...] Read more.
Chemotherapy-related cardiotoxicity (CTRTOX) is a profound and common side effect of cancer-based therapy in a subset of patients. The underlying factors and the associated mechanisms contributing to severe toxicity of the heart among these patients remain unknown. While challenges remain in accessing human subjects and their ventricular cardiomyocytes (CMs), advancements in human induced pluripotent stem cell (hiPSC)-technology-based CM differentiation protocols over the past few decades have paved the path for iPSC-based models of human cardiac diseases. Here, we offer a detailed analysis of the underlying mechanisms of CTRTOX. We also discuss the recent advances in therapeutic strategies in different animal models and clinical trials. Furthermore, we explore the prospects of iPSC-based models for identifying novel functional targets and developing safer chemotherapy regimens for cancer patients that may be beneficial for developing personalized cardioprotectants and their application in clinical practice. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

12 pages, 1410 KiB  
Article
Mutation-Specific Cardiomyocyte Lines from Patients with Fabry Disease: A Sustainable In Vitro Model to Investigate Structure, Function, and Disease Mechanisms
by Kathleen Nicholls, Andrea Wise, David Elliot, Menno ter Huurne, Maria Fuller and Sharon Ricardo
Int. J. Transl. Med. 2025, 5(2), 15; https://doi.org/10.3390/ijtm5020015 - 15 Apr 2025
Viewed by 936
Abstract
Background: Fabry disease (FD) results from pathogenic GLA variants, causing lysosomal α-galactosidase A (α-GalA) deficiency and sphingolipid ceramide trihexoside (Gb3 or THC) accumulation. Disease phenotype varies widely but cardiomyopathy is commonly life-limiting. As a multisystemic disorder, FD initiates at the cellular level; however, [...] Read more.
Background: Fabry disease (FD) results from pathogenic GLA variants, causing lysosomal α-galactosidase A (α-GalA) deficiency and sphingolipid ceramide trihexoside (Gb3 or THC) accumulation. Disease phenotype varies widely but cardiomyopathy is commonly life-limiting. As a multisystemic disorder, FD initiates at the cellular level; however, the mechanism/s underlying Gb3-induced cell dysfunction remains largely unknown. This study established an in vitro mutation-specific model of Fabry cardiomyopathy using human-induced pluripotent stem cell (iPSC)-derived cardiomyocytes to explore underlying cell pathology. Methods: Skin biopsies from consenting Fabry patients and normal control subjects were reprogrammed to iPSCs then differentiated into cardiomyocytes. The GLA mutations in Fabry cell lines were corrected using CRISP-Cas9. Phenotypic characteristics, α-Gal A activity, Gb3 accumulation, functional status, and lipid analysis were assessed. Cardiomyocytes derived from two patients with severe clinical phenotype and genotypes, GLAc.851T>C, GLAc.1193_1196del, and their respective corrected lines, GLAcorr c.851T>C, GLAcorr c.1193_1196del, were selected for further studies. Results: Cardiomyocytes derived from individuals with FD iPSCs exhibited stable expression of cardiomyocyte markers and spontaneous contraction, morphological features of FD, reduced α-Gal A activity, and accumulation of Gb3. Lipidomic profiling revealed differences in the Gb3 isoform profile between the control and FD patient iPSC-derived cardiomyocytes. Contraction strength was unchanged but relaxation after contraction was delayed, mimicking the diastolic dysfunction typical of Fabry cardiomyopathy. Conclusions: iPSC-derived cardiomyocytes provide a useful model to explore aspects of Fabry cardiomyopathy, including disruptions in sphingolipid pathways, proteomics, and multigene expression that together link genotype to phenotype. The platform potentially offers broad applicability across many genetic diseases and offers the prospect of testing and implementation of individualised therapies. Full article
Show Figures

Figure 1

27 pages, 2121 KiB  
Review
Cell Reprogramming, Transdifferentiation, and Dedifferentiation Approaches for Heart Repair
by Micael Almeida, José M. Inácio, Carlos M. Vital, Madalena R. Rodrigues, Beatriz C. Araújo and José A. Belo
Int. J. Mol. Sci. 2025, 26(7), 3063; https://doi.org/10.3390/ijms26073063 - 27 Mar 2025
Cited by 1 | Viewed by 1445
Abstract
Cardiovascular disease (CVD) remains the leading cause of death globally, with myocardial infarction (MI) being a major contributor. The current therapeutic approaches are limited in effectively regenerating damaged cardiac tissue. Up-to-date strategies for heart regeneration/reconstitution aim at cardiac remodeling through repairing the damaged [...] Read more.
Cardiovascular disease (CVD) remains the leading cause of death globally, with myocardial infarction (MI) being a major contributor. The current therapeutic approaches are limited in effectively regenerating damaged cardiac tissue. Up-to-date strategies for heart regeneration/reconstitution aim at cardiac remodeling through repairing the damaged tissue with an external cell source or by stimulating the existing cells to proliferate and repopulate the compromised area. Cell reprogramming is addressed to this challenge as a promising solution, converting fibroblasts and other cell types into functional cardiomyocytes, either by reverting cells to a pluripotent state or by directly switching cell lineage. Several strategies such as gene editing and the application of miRNA and small molecules have been explored for their potential to enhance cardiac regeneration. Those strategies take advantage of cell plasticity by introducing reprogramming factors that regress cell maturity in vitro, allowing for their later differentiation and thus endorsing cell transplantation, or promote in situ cell proliferation, leveraged by scaffolds embedded with pro-regenerative factors promoting efficient heart restoration. Despite notable advancements, important challenges persist, including low reprogramming efficiency, cell maturation limitations, and safety concerns in clinical applications. Nonetheless, integrating these innovative approaches offers a promising alternative for restoring cardiac function and reducing the dependency on full heart transplants. Full article
Show Figures

Figure 1

23 pages, 1871 KiB  
Review
Microgravity and Cellular Biology: Insights into Cellular Responses and Implications for Human Health
by Nelson Adolfo López Garzón, María Virginia Pinzón-Fernández, Jhan S. Saavedra T., Humberto A. Nati-Castillo, Marlon Arias-Intriago, Camila Salazar-Santoliva and Juan S. Izquierdo-Condoy
Int. J. Mol. Sci. 2025, 26(7), 3058; https://doi.org/10.3390/ijms26073058 - 27 Mar 2025
Cited by 2 | Viewed by 1873
Abstract
Microgravity, defined by minimal gravitational forces, represents a unique environment that profoundly influences biological systems, including human cells. This review examines the effects of microgravity on biological processes and their implications for human health. Microgravity significantly impacts the immune system by disrupting key [...] Read more.
Microgravity, defined by minimal gravitational forces, represents a unique environment that profoundly influences biological systems, including human cells. This review examines the effects of microgravity on biological processes and their implications for human health. Microgravity significantly impacts the immune system by disrupting key mechanisms, such as T cell activation, cytokine production, and macrophage differentiation, leading to increased susceptibility to infections. In cancer biology, it promotes the formation of spheroids in cancer stem cells and thyroid cancer cells, which closely mimic in vivo tumor dynamics, providing novel insights for oncology research. Additionally, microgravity enhances tissue regeneration by modulating critical pathways, including Hippo and PI3K-Akt, thereby improving stem cell differentiation into hematopoietic and cardiomyocyte lineages. At the organ level, microgravity induces notable changes in hepatic metabolism, endothelial function, and bone mechanotransduction, contributing to lipid dysregulation, vascular remodeling, and accelerated bone loss. Notably, cardiomyocytes derived from human pluripotent stem cells and cultured under microgravity exhibit enhanced mitochondrial biogenesis, improved calcium handling, and advanced structural maturation, including increased sarcomere length and nuclear eccentricity. These advancements enable the development of functional cardiomyocytes, presenting promising therapeutic opportunities for treating cardiac diseases, such as myocardial infarctions. These findings underscore the dual implications of microgravity for space medicine and terrestrial health. They highlight its potential to drive advances in regenerative therapies, oncology, and immunological interventions. Continued research into the biological effects of microgravity is essential for protecting astronaut health during prolonged space missions and fostering biomedical innovations with transformative applications on Earth. Full article
Show Figures

Figure 1

29 pages, 1028 KiB  
Review
Advances in Conductive Biomaterials for Cardiac Tissue Engineering: Design, Fabrication, and Functional Integration
by Tabrej Khan, Gayathri Vadivel, Kalaivani Ayyasamy, Gowtham Murugesan and Tamer A. Sebaey
Polymers 2025, 17(5), 620; https://doi.org/10.3390/polym17050620 - 26 Feb 2025
Viewed by 2693
Abstract
Heart failure functions as one of the leading global causes of death because it falls under the cardiovascular disease categories. Cardiac tissue engineering advances by developing new tissues to rebuild heart functions in individuals with damaged heart structures as it gives medical treatment [...] Read more.
Heart failure functions as one of the leading global causes of death because it falls under the cardiovascular disease categories. Cardiac tissue engineering advances by developing new tissues to rebuild heart functions in individuals with damaged heart structures as it gives medical treatment possibilities to patients reaching their final stage. Most of the heart tissue consists of cardiomyocytes which make up between 80 to 90 percent of the total organ space. The cardiomyocytes retain their specialized cell structure which includes elongation, but they align to produce contractions as they span into length. After myocardial infarction, doctors need elastic soft platforms to heal the heart tissue because they mimic its natural attributes. Special consideration must be paid to the material selection for appropriate mechanical properties, given that different substances have separate qualities. Stem cell survival becomes higher, and cell differentiation develops more efficiently when a proper scaffold design is implemented, thus enabling tissue repair. Conductive biomaterials demonstrate the best candidate status for cardiac tissue engineering due to their ability to both convey electrical signals and boost biological actions as well as promote cellular communication. Scientists conduct life science research on stem cells because the cells present unique characteristics. Biomaterials with conductive properties within cardiac tissue engineering help the body recover heart tissue while improving the functionality of damaged structures in the myocardium. This article analyzes various conductive biomaterials used in biomedical practices for cardiac tissue healing applications. Full article
Show Figures

Figure 1

22 pages, 27687 KiB  
Article
RNA-Binding Protein Signature in Proliferative Cardiomyocytes: A Cross-Species Meta-Analysis from Mouse, Pig, and Human Transcriptomic Profiling Data
by Thanh Nguyen, Kaili Hao, Yuji Nakada, Bijay Guragain, Peng Yao and Jianyi Zhang
Biomolecules 2025, 15(2), 310; https://doi.org/10.3390/biom15020310 - 19 Feb 2025
Cited by 1 | Viewed by 1360
Abstract
In mammals, because cardiomyocytes withdraw from cell-cycle activities shortly after birth, the heart cannot repair the damage caused by a myocardial injury; thus, understanding how cardiomyocytes proliferate is among the most important topics in cardiovascular sciences. In newborn neonatal mammals, when a left [...] Read more.
In mammals, because cardiomyocytes withdraw from cell-cycle activities shortly after birth, the heart cannot repair the damage caused by a myocardial injury; thus, understanding how cardiomyocytes proliferate is among the most important topics in cardiovascular sciences. In newborn neonatal mammals, when a left ventricular injury is applied in hearts earlier than postnatal day 7, the cardiomyocytes actively proliferate and regenerate lost myocardium in the following weeks. The regulators promoting cardiomyocyte proliferation were discovered by analyzing transcriptomic data generated from models. Most of these regulators support the mRNA production of cell-cycle machinery, yet the mRNA requires translation into functional proteins under the regulation of RNA-binding proteins (RBPs). In this work, we performed a meta-analysis to study the relationship between RBP expression and cardiomyocyte proliferation. To identify RBPs associated with mouse and pig cardiomyocyte proliferation, the single-nuclei RNA sequencing (snRNA-seq) data from regenerating mouse and pig hearts were reanalyzed via an Autoencoder focusing on RBP expression. We also generated and analyzed new bulk RNA-seq from two human-induced pluripotent stem cell-derived (hiPSC) cardiomyocyte (hiPSC-CM) cell lines; the first cell line was harvested sixteen days after differentiation, when the cells still actively proliferated, and the second cell line was harvested one hundred and forty days after differentiation, when the cells ceased cell cycle activity. Then, the RBP associated with mouse, pig, and hiPSC-CM were compared across species. Twenty-one RBPs were found to be consistently upregulated, and six RBPs were downregulated in proliferating mouse, pig, and hiPSC-derived cardiomyocytes. Among upregulated RBPs across species, an immunofluorescence-based imaging analysis validated the significant increase in the proteins of DHX9, PTBP3, HNRNPUL1, and DDX6 in pig hearts with proliferating CMs. This meta-analysis in all species demonstrated a strong relationship between RBP expression and cardiomyocyte proliferation. Full article
Show Figures

Figure 1

32 pages, 29372 KiB  
Article
Transcriptome-Wide Insights: Neonatal Lactose Intolerance Promotes Telomere Damage, Senescence, and Cardiomyopathy in Adult Rat Heart
by Olga V. Anatskaya, Sergei V. Ponomartsev, Artem U. Elmuratov and Alexander E. Vinogradov
Int. J. Mol. Sci. 2025, 26(4), 1584; https://doi.org/10.3390/ijms26041584 - 13 Feb 2025
Viewed by 3961
Abstract
Cardiovascular diseases (CVD) are the primary cause of mortality globally. A significant aspect of CVD involves their association with aging and susceptibility to neonatal programming. These factors suggest that adverse conditions during neonatal development can disrupt cardiomyocyte differentiation, thereby leading to heart dysfunction. [...] Read more.
Cardiovascular diseases (CVD) are the primary cause of mortality globally. A significant aspect of CVD involves their association with aging and susceptibility to neonatal programming. These factors suggest that adverse conditions during neonatal development can disrupt cardiomyocyte differentiation, thereby leading to heart dysfunction. This study focuses on the long-term effects of inflammatory and oxidative stress due to neonatal lactose intolerance (NLI) on cardiomyocyte transcriptome and phenotype. Our recent bioinformatic study focused on toggle genes indicated that NLI correlates with the switch off of some genes in thyroid hormone, calcium, and antioxidant signaling pathways, alongside the switch-on/off genes involved in DNA damage response and inflammation. In the presented study, we evaluated cardiomyocyte ploidy in different regions of the left ventricle (LV), complemented by a transcriptomic analysis of genes with quantitative (gradual) difference in expression. Cytophotometric and morphologic analyses of LV cardiomyocytes identified hyperpolyploidy and bridges between nuclei suggesting telomere fusion. Transcriptomic profiling highlighted telomere damage, aging, and chromatin decompaction, along with the suppression of pathways governing muscle contraction and energy metabolism. Echocardiography revealed statistically significant LV dilation and a decrease in ejection fraction. The estimation of survival rates indicated that NLI shortened the median lifespan by approximately 18% (p < 0.0001) compared with the control. Altogether, these findings suggest that NLI may increase susceptibility to cardiovascular diseases by accelerating aging due to oxidative stress and increased telomere DNA damage, leading to hyperpolyploidization and reduced cardiac contractile function. Collectively, our data emphasize the importance of the early identification and management of neonatal inflammatory and metabolic stressors, such as NLI, to mitigate long-term cardiovascular risks. Full article
Show Figures

Figure 1

17 pages, 4074 KiB  
Article
A Platform Integrating Biophysical and Biochemical Stimuli to Enhance Differentiation and Maturation of Cardiomyocyte Subtypes Derived from Human Induced Pluripotent Stem Cells
by Zhonggang Feng, Kota Sawada, Iori Ando, Riku Yoshinari, Daisuke Sato and Tadashi Kosawada
J. Cardiovasc. Dev. Dis. 2025, 12(2), 56; https://doi.org/10.3390/jcdd12020056 - 4 Feb 2025
Viewed by 1025
Abstract
To enhance the differentiation and maturation of cardiomyocytes derived from human induced pluripotent stem cells, we developed a bioreactor system that simultaneously imposes biophysical and biochemical stimuli on these committed cardiomyocytes. The cells were cultured within biohydrogels composed of the extracellular matrix extracted [...] Read more.
To enhance the differentiation and maturation of cardiomyocytes derived from human induced pluripotent stem cells, we developed a bioreactor system that simultaneously imposes biophysical and biochemical stimuli on these committed cardiomyocytes. The cells were cultured within biohydrogels composed of the extracellular matrix extracted from goat ventricles and purchased rat-origin collagen, which were housed in the elastic PDMS culture chambers of the bioreactor. Elastic and flexible electrodes composed of PEDOT/PSS, latex, and graphene flakes were embedded in the hydrogels and chamber walls, allowing cyclic stretch and electrical pulses to be simultaneously and coordinately applied to the cultured cells. Furthermore, a dynamic analysis method employing the transverse forced oscillation theory of a cantilever was used to analyze and discriminate the subtype-specific beating behavior of the cardiomyocytes. It was found that myosin light chain 2v (MLC2v), a ventricular cell marker, was primarily upregulated in cells aggregated on the (+) electrode side, while cardiomyocytes with faint MLC2v but strong cardiac troponin T (cTNT) expression aggregated at the ground electrode (GND) side. mRNA analysis using rtPCR and the gel beating dynamics further suggested a subtype deviation on the different electrode sides. This study demonstrated the potential of our bioreactor system in enhancing cardiac differentiation and maturation, and it showed an intriguing phenomenon of cardiomyocyte subtype aggregation on different electrodes, which may be developed into a new method to enhance the maturation and separation of cardiomyocyte subtypes. Full article
(This article belongs to the Section Cardiac Development and Regeneration)
Show Figures

Figure 1

9 pages, 947 KiB  
Article
Differentially Expressed Genes in Cardiomyocytes of the First Camelized Mouse Model, Nrapc.255ins78 Mouse
by Sung-Yeon Lee, Byeonghwi Lim, Bo-Young Lee, Goo Jang, Jung-Seok Choi, Xiang-Shun Cui and Kwan-Suk Kim
Genes 2025, 16(2), 142; https://doi.org/10.3390/genes16020142 - 24 Jan 2025
Viewed by 1272
Abstract
Background/Objectives: The first camelized mouse model (Nrapc.255ins78) was developed to investigate the mechanisms underlying camels’ adaptation to extreme environments. Previous studies demonstrated that these mice exhibit a cold-resistant phenotype, characterized by increased expression of inflammatory cytokine-related genes in the heart [...] Read more.
Background/Objectives: The first camelized mouse model (Nrapc.255ins78) was developed to investigate the mechanisms underlying camels’ adaptation to extreme environments. Previous studies demonstrated that these mice exhibit a cold-resistant phenotype, characterized by increased expression of inflammatory cytokine-related genes in the heart under cold stress. Nebulin-related anchoring protein (NRAP) plays a critical role in organizing myofibrils during cardiomyocyte development. This study builds on prior research by analyzing the heart transcriptomes of Nrapc.255ins78 mice under non-stress conditions to explore the origins of inflammatory cytokine responses during cold exposure. Methods: RNA sequencing was performed on the hearts of 12-week-old male and female Nrapc.255ins78 and wild-type control mice. Results: Differential expression analysis identified 25 genes, including 12 associated with cell cycle and division, all consistently downregulated in Nrapc.255ins78. Notably, the calcium and integrin-binding protein gene (Cib3) was significantly upregulated (FDR < 0.05; p < 0.001). Conclusions: These differentially expressed genes suggest altered calcium dynamics in cardiomyocytes and mechanisms for maintaining homeostasis, supporting the hypothesis that inflammatory cytokines during cold exposure may represent an adaptive response. These findings provide valuable insights into the genetic mechanisms of temperature adaptation in camels and highlight potential pathways for enhancing stress resistance in other mammals. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

Back to TopTop