Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 202
Filtrer
1.
J Mol Cell Cardiol ; 196: 52-70, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39222876

RÉSUMÉ

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are advancing cardiovascular development and disease modeling, drug testing, and regenerative therapies. However, hPSC-CM production is hindered by significant variability in the differentiation process. Establishment of early quality markers to monitor lineage progression and predict terminal differentiation outcomes would address this robustness and reproducibility roadblock in hPSC-CM production. An integrated transcriptomic and epigenomic analysis assesses how attributes of the cardiac progenitor cell (CPC) affect CM differentiation outcome. Resulting analysis identifies predictive markers of CPCs that give rise to high purity CM batches, including TTN, TRIM55, DGKI, MEF2C, MAB21L2, MYL7, LDB3, SLC7A11, and CALD1. Predictive models developed from these genes provide high accuracy in determining terminal CM purities at the CPC stage. Further, insights into mechanisms of batch failure and dominant non-CM cell types generated in failed batches are elucidated. Namely EMT, MAPK, and WNT signaling emerge as significant drivers of batch divergence, giving rise to off-target populations of fibroblasts/mural cells, skeletal myocytes, epicardial cells, and a non-CPC SLC7A11+ subpopulation. This study demonstrates how integrated multi-omic analysis of progenitor cells can identify quality attributes of that progenitor and predict differentiation outcomes, thereby improving differentiation protocols and increasing process robustness.

2.
Methods Mol Biol ; 2835: 147-154, 2024.
Article de Anglais | MEDLINE | ID: mdl-39105913

RÉSUMÉ

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.


Sujet(s)
Cellules souches , Animaux , Cellules souches/métabolisme , Cellules souches/cytologie , Glycolyse , Myocarde/métabolisme , Myocarde/cytologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/cytologie , Souris , Humains , Différenciation cellulaire , Phosphorylation oxydative , Prolifération cellulaire , Séparation cellulaire/méthodes
3.
Methods Mol Biol ; 2835: 269-275, 2024.
Article de Anglais | MEDLINE | ID: mdl-39105922

RÉSUMÉ

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.


Sujet(s)
Techniques de culture cellulaire , Conductivité électrique , Polymères , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Structures d'échafaudage tissulaires/composition chimique , Ingénierie tissulaire/méthodes , Polymères/composition chimique , Techniques de culture cellulaire/méthodes , Pyrroles/composition chimique , Animaux , Humains , Matériaux biocompatibles/composition chimique , Cellules cultivées , Cellules souches/cytologie , Dérivés de l'aniline/composition chimique , Myocytes cardiaques/cytologie , Stéréolithographie
4.
Adv Exp Med Biol ; 1441: 875-884, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884756

RÉSUMÉ

Tricuspid atresia (TA) is a rare congenital heart condition that presents with a complete absence of the right atrioventricular valve. Because of the rarity of familial and/or isolated cases of TA, little is known about the potential genetic abnormalities contributing to this condition. Potential responsible chromosomal abnormalities were identified in exploratory studies and include deletions in 22q11, 4q31, 8p23, and 3p as well as trisomies 13 and 18. In parallel, potential culprit genes include the ZFPM2, HEY2, NFATC1, NKX2-5, MYH6, and KLF13 genes. The aim of this chapter is to expose the genetic components that are potentially involved in the pathogenesis of TA in humans. The large variability in phenotypes and genotypes among cases of TA suggests a genetic network that involves many components yet to be unraveled.


Sujet(s)
Atrésie tricuspide , Humains , Aberrations des chromosomes , Phénotype , Atrésie tricuspide/génétique , Cœur univentriculaire/génétique
5.
Stem Cell Res ; 79: 103476, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38941882

RÉSUMÉ

Cardiovascular disease remains a global health concern. Stem cell therapy utilizing human cardiac progenitor cells (hCPCs) shows promise in treating cardiac vascular disease. However, limited availability and senescence of hCPCs hinder their widespread use. To address these challenges, researchers are exploring innovative approaches. In this study, a bioengineered cell culture plate was developed to mimic the natural cardiac tissue microenvironment. It was coated with a combination of extracellular matrix (ECM) peptide motifs and mussel adhesive protein (MAP). The selected ECM peptide motifs, derived from fibronectin and vitronectin, play crucial roles in hCPCs. Results revealed that the Fibro-P and Vitro-P coated plates significantly improved hCPC adhesion, proliferation, migration, and differentiation compared to uncoated plates. Additionally, long-term culture on the coated plates delayed cellular senescence and maintained hCPC stemness. These enhancements were attributed to the activation of integrin downstream signaling pathways. The findings suggest that the engineered ECM peptide motif-MAP-coated plates hold potential for enhancing the therapeutic efficacy of stem cell-based therapies in cardiac tissue engineering and regenerative medicine.


Sujet(s)
Fibronectines , Cellules souches , Vitronectine , Vitronectine/métabolisme , Humains , Fibronectines/métabolisme , Cellules souches/cytologie , Cellules souches/métabolisme , Différenciation cellulaire , Techniques de culture cellulaire/méthodes , Cellules cultivées , Prolifération cellulaire , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Adhérence cellulaire , Peptides
6.
Adv Exp Med Biol ; 1441: 77-85, 2024.
Article de Anglais | MEDLINE | ID: mdl-38884705

RÉSUMÉ

The major events of cardiac development, including early heart formation, chamber morphogenesis and septation, and conduction system and coronary artery development, are briefly reviewed together with a short introduction to the animal species commonly used to study heart development and model congenital heart defects (CHDs).


Sujet(s)
Modèles animaux de maladie humaine , Cardiopathies congénitales , Coeur , Animaux , Cardiopathies congénitales/physiopathologie , Cardiopathies congénitales/anatomopathologie , Coeur/embryologie , Coeur/physiopathologie , Coeur/croissance et développement , Humains , Souris , Morphogenèse
7.
Curr Res Toxicol ; 6: 100167, 2024.
Article de Anglais | MEDLINE | ID: mdl-38659494

RÉSUMÉ

Sunitinib malate is known to cause cardiotoxicity in a sub-population of patients, with heart failure seen in more severe cases. Cardiac progenitor cells (CPCs) have been identified in adult human myocardium and contribute to overall tissue maintenance, with previous work identifying negative impacts of sunitinib on these cells. This study aimed to characterise the toxic effects of sunitinib in human CPCs, applying sunitinib concentrations equivalent to clinical plasma levels to these cells in vitro. Cell viability was reduced by 26.5 ± 6.6 % by 2 µM sunitinib for 24 h (p < 0.01); this concentration also induced fold-change increases in gene expression of: calpain (3.1 ± 0.73, p < 0.05), FAS (2.3 ± 0.8, p < 0.05) and BAX (1.9 ± 0.2, p < 0.05), and a decrease in BCL-2 (3.5 ± 0.0, p < 0.001), vs. control (1.0 ± 0.0). This was affirmed by sunitinib inducing fold changes in protein expression of: calpain-1 (2.5 ± 0.5, p < 0.05); FAS receptor (2.1 ± 0.2, p < 0.05) and BAX (2.1 ± 0.2, p < 0.05) vs. control (1.0 ± 0.0). These results indicated that sunitinib induced apoptosis in CPCs, but negative annexin V staining and lack of protection by caspase inhibitors indicated this was not the cell death pathway activated. Further investigation found sunitinib was concentrated in the lysosomes and autophagosomes within CPCs, but did not induce accumulation of acidic organelles. In conclusion, these data confirm that cell death is caused by sunitinib in CPCs at concentrations equivalent to clinical plasma levels, inducing cell death pathway signals that lead to non-apoptotic cell death.

8.
Cell Biosci ; 14(1): 30, 2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38444042

RÉSUMÉ

Heart failure (HF) is an end-stage of many cardiac diseases and one of the main causes of death worldwide. The current management of this disease remains suboptimal. The adult mammalian heart was considered a post-mitotic organ. However, several reports suggest that it may possess modest regenerative potential. Adult cardiac progenitor cells (CPCs), the main players in the cardiac regeneration, constitute, as it may seem, a heterogenous group of cells, which remain quiescent in physiological conditions and become activated after an injury, contributing to cardiomyocytes renewal. They can mediate their beneficial effects through direct differentiation into cardiac cells and activation of resident stem cells but majorly do so through paracrine release of factors. CPCs can secrete cytokines, chemokines, and growth factors as well as exosomes, rich in proteins, lipids and non-coding RNAs, such as miRNAs and YRNAs, which contribute to reparation of myocardium by promoting angiogenesis, cardioprotection, cardiomyogenesis, anti-fibrotic activity, and by immune modulation. Preclinical studies assessing cardiac progenitor cells and cardiac progenitor cells-derived exosomes on damaged myocardium show that administration of cardiac progenitor cells-derived exosomes can mimic effects of cell transplantation. Exosomes may become new promising therapeutic strategy for heart regeneration nevertheless there are still several limitations as to their use in the clinic. Key questions regarding their dosage, safety, specificity, pharmacokinetics, pharmacodynamics and route of administration remain outstanding. There are still gaps in the knowledge on basic biology of exosomes and filling them will bring as closer to translation into clinic.

9.
C R Biol ; 347: 9-18, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38488639

RÉSUMÉ

Congenital heart defects (CHD) affect 1 in 100 live births and result from defects in cardiac development. Growth of the early heart tube occurs by the progressive addition of second heart field (SHF) progenitor cells to the cardiac poles. The SHF gives rise to ventricular septal, right ventricular and outflow tract myocardium at the arterial pole, and atrial, including atrial septal myocardium, at the venous pole. SHF deployment creates the template for subsequent cardiac septation and has been implicated in cardiac looping and in orchestrating outflow tract development with neural crest cells. Genetic or environmental perturbation of SHF deployment thus underlies a spectrum of common forms of CHD affecting conotruncal and septal morphogenesis. Here we review the major properties of SHF cells as well as recent insights into the developmental programs that drive normal cardiac progenitor cell addition and the origins of CHD.


Les malformations cardiaques congénitales touchent 1 naissance sur 100 et résultent d'anomalies du développement cardiaque. La croissance du tube cardiaque précoce se produit par l'ajout progressif de cellules progénitrices du second champ cardiaque (SHF) aux pôles cardiaques. Le SHF contribue au myocarde septal ventriculaire, au myocarde ventriculaire droit et au myocarde de la voie de sortie au pôle artériel, et au myocarde auriculaire, y compris le myocarde septal auriculaire, au pôle veineux. Le déploiement du SHF est essentiel pour la septation cardiaque et a été impliqué dans la formation du boucle cardiaque et, avec les cellules de la crête neurale, dans l'orchestration du développement de la voie efférente. Perturbation génétique ou environnementale du déploiement du SHF est donc à l'origine d'un spectre de formes communes de maladies cardiaques congénitales affectant la morphogenèse conotroncale et septale. Ici, nous passons en revue les principales propriétés des cellules du SHF ainsi que les découvertes récentes sur les programmes de développement qui contrôlent l'ajout de cellules progénitrices cardiaques ainsi que les origines des malformations cardiaques congénitales.


Sujet(s)
Cardiopathies congénitales , Coeur , Humains , Cardiopathies congénitales/génétique , Myocarde , Cellules souches , Morphogenèse
11.
Methods Mol Biol ; 2729: 303-330, 2024.
Article de Anglais | MEDLINE | ID: mdl-38006504

RÉSUMÉ

Noninvasive long-term imaging of therapeutic cells in preclinical models can be achieved through introducing a reporter gene into the cells of interest. Despite important recent developments such as gene editing, cell engineering based on lentiviruses remains a mainstream tool for gene transfer applicable to a variety of different cell types.In this chapter, we describe how to use lentivirus-based genetic engineering to render different candidate cell therapies in vivo traceable by radionuclide imaging. We illustrate this reporter gene technology using the sodium iodide symporter (NIS), which is compatible with both positron emission tomography (PET) and single-photon emission computed tomography (SPECT). For preclinical experimentation, we fused NIS with a suitable fluorescent protein such as monomeric GFP or RFP to streamline cell line generation and downstream analyses of ex vivo tissue samples. We present protocols for reporter gene engineering of human cardiac progenitor cells, regulatory T cells, and effector T cells as well as for the characterization experiments required to validate NIS-fluorescent protein reporter function in these candidate therapeutic cells.


Sujet(s)
Tomographie par émission de positons , Symporteurs , Humains , Tomographie par émission de positons/méthodes , Symporteurs/génétique , Symporteurs/métabolisme , Tomographie par émission monophotonique , Génie génétique
12.
BMC Cancer ; 23(1): 1245, 2023 Dec 18.
Article de Anglais | MEDLINE | ID: mdl-38110859

RÉSUMÉ

BACKGROUND: Cardiac Myxoma is a primary tumor of heart. Its origins, rarity of the occurrence of primary cardiac tumors and how it may be related to limited cardiac regenerative potential, are not yet entirely known. This study investigates the key cardiac genes/ transcription factors (TFs) and signaling pathways to understand these important questions. METHODS: Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving cardiac myxoma, cardiac genes/TFs/signaling pathways and their roles in cardiogenesis, proliferation, differentiation, key interactions and tumorigenesis, with focus on cardiomyocytes. RESULTS: The cardiac genetic landscape is governed by a very tight control between proliferation and differentiation-related genes/TFs/pathways. Cardiac myxoma originates possibly as a consequence of dysregulations in the gene expression of differentiation regulators including Tbx5, GATA4, HAND1/2, MYOCD, HOPX, BMPs. Such dysregulations switch the expression of cardiomyocytes into progenitor-like state in cardiac myxoma development by dysregulating Isl1, Baf60 complex, Wnt, FGF, Notch, Mef2c and others. The Nkx2-5 and MSX2 contribute predominantly to both proliferation and differentiation of Cardiac Progenitor Cells (CPCs), may possibly serve roles based on the microenvironment and the direction of cell circuitry in cardiac tumorigenesis. The Nkx2-5 in cardiac myxoma may serve to limit progression of tumorigenesis as it has massive control over the proliferation of CPCs. The cardiac cell type-specific genetic programming plays governing role in controlling the tumorigenesis and regenerative potential. CONCLUSION: The cardiomyocytes have very limited proliferative and regenerative potential. They survive for long periods of time and tightly maintain the gene expression of differentiation genes such as Tbx5, GATA4 that interact with tumor suppressors (TS) and exert TS like effect. The total effect such gene expression exerts is responsible for the rare occurrence and benign nature of primary cardiac tumors. This prevents the progression of tumorigenesis. But this also limits the regenerative and proliferative potential of cardiomyocytes. Cardiac Myxoma develops as a consequence of dysregulations in these key genes which revert the cells towards progenitor-like state, hallmark of CM. The CM development in carney complex also signifies the role of TS in cardiac cells.


Sujet(s)
Tumeurs du coeur , Myxome , Humains , Facteurs de transcription/métabolisme , Myocytes cardiaques/physiologie , Différenciation cellulaire/génétique , Tumeurs du coeur/génétique , Tumeurs du coeur/anatomopathologie , Myxome/génétique , Myxome/métabolisme , Myxome/anatomopathologie , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Microenvironnement tumoral
13.
Heliyon ; 9(11): e21268, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37954289

RÉSUMÉ

Background: Subpopulations of myocardial c-kitpos cells have the ability to stimulate regeneration in ischemic heart disease by paracrine effects. The left atrial appendage (LAA), which is easy accessible during cardiac surgery, may represent a perfect source for c-kitpos cell extraction for autologous cell therapies in the living human. So far, frequency and distribution of c-kitpos cells in LAA are unknown. Methods: LAAs of patients who underwent cardiac surgery due to coronary artery disease (coronary artery bypass graft, CABG), valvular heart disease or both and of two body donors were examined. Tissue was fixed in 4 % paraformaldehyde, embedded in paraffin, dissected in consecutive sections and stained for c-kitpos cells. In parallel, grade of fibrosis, amount of fat per section and cells positive for mast cell tryptase were examined. Results: We collected 27 LAAs (37.0 % female, mean left ventricular ejection fraction 50.4 %, 63.0 % persistent atrial fibrillation (AF)). Most of the patients underwent combined CABG and valve surgery (51.9 %). C-kitpos cells were detected in 3 different regions: A) Attached to the epicardial fat layer, B) close to vascular structures and C) between cardiomyocytes. C-kitpos cells ranged from 0.05 c-kitpos cells per mm2 to 67.5 c-kitpos cells per mm2. We found no association between number of c-kitpos cells and type of AF, amount of fibrosis or amount of fat. Up to 72 % of c-kitpos cells also showed a positive staining for mast cell tryptase. Conclusion: C-kitpos cells are frequent in LAAs of cardiovascular patients with a rather homogenous distribution throughout the LAA. The LAA can therefore be considered as a source for extraction of a reasonable quantity of autologous cardiac progenitor cells in the living human patient.

14.
Int J Mol Sci ; 24(15)2023 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-37569446

RÉSUMÉ

This study investigated the protective effect of glutathione (GSH), an antioxidant drug, against doxorubicin (DOX)-induced cardiotoxicity. Human cardiac progenitor cells (hCPCs) treated with DOX (250 to 500 nM) showed increased viability and reduced ROS generation and apoptosis with GSH treatment (0.1 to 1 mM) for 24 h. In contrast to the 500 nM DOX group, pERK levels were restored in the group co-treated with GSH and suppression of ERK signaling improved hCPCs' survival. Similarly to the previous results, the reduced potency of hCPCs in the 100 nM DOX group, which did not affect cell viability, was ameliorated by co-treatment with GSH (0.1 to 1 mM). Furthermore, GSH was protected against DOX-induced cardiotoxicity in the in vivo model (DOX 20 mg/kg, GSH 100 mg/kg). These results suggest that GSH is a potential therapeutic strategy for DOX-induced cardiotoxicity, which performs its function via ROS reduction and pERK signal regulation.

15.
Int J Mol Sci ; 24(12)2023 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-37373444

RÉSUMÉ

Ischemic heart disease (IHD) is the leading cause of heart failure (HF) and is a significant cause of morbidity and mortality globally. An ischemic event induces cardiomyocyte death, and the ability for the adult heart to repair itself is challenged by the limited proliferative capacity of resident cardiomyocytes. Intriguingly, changes in metabolic substrate utilisation at birth coincide with the terminal differentiation and reduced proliferation of cardiomyocytes, which argues for a role of cardiac metabolism in heart regeneration. As such, strategies aimed at modulating this metabolism-proliferation axis could, in theory, promote heart regeneration in the setting of IHD. However, the lack of mechanistic understanding of these cellular processes has made it challenging to develop therapeutic modalities that can effectively promote regeneration. Here, we review the role of metabolic substrates and mitochondria in heart regeneration, and discuss potential targets aimed at promoting cardiomyocyte cell cycle re-entry. While advances in cardiovascular therapies have reduced IHD-related deaths, this has resulted in a substantial increase in HF cases. A comprehensive understanding of the interplay between cardiac metabolism and heart regeneration could facilitate the discovery of novel therapeutic targets to repair the damaged heart and reduce risk of HF in patients with IHD.


Sujet(s)
Défaillance cardiaque , Ischémie myocardique , Nouveau-né , Humains , Coeur , Myocytes cardiaques/métabolisme , Ischémie myocardique/métabolisme , Défaillance cardiaque/métabolisme , Prolifération cellulaire
16.
Front Cell Dev Biol ; 11: 1111684, 2023.
Article de Anglais | MEDLINE | ID: mdl-37261075

RÉSUMÉ

Domestic pigs (Sus scrofa) share many genetic, anatomical, and physiological traits with humans and therefore constitute an excellent preclinical animal model. Fundamental understanding of the cellular and molecular processes governing early porcine cardiogenesis is critical for developing advanced porcine models used for the study of heart diseases and new regenerative therapies. Here, we provide a detailed characterization of porcine cardiogenesis based on fetal porcine hearts at various developmental stages and cardiac cells derived from porcine expanded pluripotent stem cells (pEPSCs), i.e., stem cells having the potential to give rise to both embryonic and extraembryonic tissue. We notably demonstrate for the first time that pEPSCs can differentiate into cardiovascular progenitor cells (CPCs), functional cardiomyocytes (CMs), epicardial cells and epicardial-derived cells (EPDCs) in vitro. Furthermore, we present an enhanced system for whole-embryo culture which allows continuous ex utero development of porcine post-implantation embryos from the cardiac crescent stage (ED14) up to the cardiac looping (ED17) stage. These new techniques provide a versatile platform for studying porcine cardiac development and disease modeling.

17.
Cells ; 12(8)2023 04 15.
Article de Anglais | MEDLINE | ID: mdl-37190075

RÉSUMÉ

Cardiac diseases are the foremost cause of morbidity and mortality worldwide. The heart has limited regenerative potential; therefore, lost cardiac tissue cannot be replenished after cardiac injury. Conventional therapies are unable to restore functional cardiac tissue. In recent decades, much attention has been paid to regenerative medicine to overcome this issue. Direct reprogramming is a promising therapeutic approach in regenerative cardiac medicine that has the potential to provide in situ cardiac regeneration. It consists of direct cell fate conversion of one cell type into another, avoiding transition through an intermediary pluripotent state. In injured cardiac tissue, this strategy directs transdifferentiation of resident non-myocyte cells (NMCs) into mature functional cardiac cells that help to restore the native tissue. Over the years, developments in reprogramming methods have suggested that regulation of several intrinsic factors in NMCs can help to achieve in situ direct cardiac reprogramming. Among NMCs, endogenous cardiac fibroblasts have been studied for their potential to be directly reprogrammed into both induced cardiomyocytes and induced cardiac progenitor cells, while pericytes can transdifferentiate towards endothelial cells and smooth muscle cells. This strategy has been indicated to improve heart function and reduce fibrosis after cardiac injury in preclinical models. This review summarizes the recent updates and progress in direct cardiac reprogramming of resident NMCs for in situ cardiac regeneration.


Sujet(s)
Transdifférenciation cellulaire , Techniques de reprogrammation cellulaire , Reprogrammation cellulaire , Fibroblastes , Cardiopathies , Coeur , Péricytes , Régénération , Coeur/physiologie , Cardiopathies/thérapie , Fibroblastes/cytologie , Fibroblastes/physiologie , Myocytes cardiaques/cytologie , Myocytes cardiaques/physiologie , Péricytes/cytologie , Péricytes/physiologie , Cellules endothéliales/cytologie , Cellules endothéliales/physiologie , Humains , Animaux
18.
Cell Transplant ; 32: 9636897231174078, 2023.
Article de Anglais | MEDLINE | ID: mdl-37191272

RÉSUMÉ

Many studies have explored cardiac progenitor cell (CPC) therapy for heart disease. However, optimal scaffolds are needed to ensure the engraftment of transplanted cells. We produced a three-dimensional hydrogel scaffold (CPC-PRGmx) in which high-viability CPCs were cultured for up to 8 weeks. CPC-PRGmx contained an RGD peptide-conjugated self-assembling peptide with insulin-like growth factor-1 (IGF-1). Immediately after creating myocardial infarction (MI), we transplanted CPC-PRGmx into the pericardial space on to the surface of the MI area. Four weeks after transplantation, red fluorescent protein-expressing CPCs and in situ hybridization analysis in sex-mismatched transplantations revealed the engraftment of CPCs in the transplanted scaffold (which was cellularized with host cells). The average scar area of the CPC-PRGmx-treated group was significantly smaller than that of the non-treated group (CPC-PRGmx-treated group = 46 ± 5.1%, non-treated MI group = 59 ± 4.5%; p < 0.05). Echocardiography showed that the transplantation of CPC-PRGmx improved cardiac function and attenuated cardiac remodeling after MI. The transplantation of CPCs-PRGmx promoted angiogenesis and inhibited apoptosis, compared to the untreated MI group. CPCs-PRGmx secreted more vascular endothelial growth factor than CPCs cultured on two-dimensional dishes. Genetic fate mapping revealed that CPC-PRGmx-treated mice had more regenerated cardiomyocytes than non-treated mice in the MI area (CPC-PRGmx-treated group = 0.98 ± 0.25%, non-treated MI group = 0.25 ± 0.04%; p < 0.05). Our findings reveal the therapeutic potential of epicardial-transplanted CPC-PRGmx. Its beneficial effects may be mediated by sustainable cell viability, paracrine function, and the enhancement of de novo cardiomyogenesis.


Sujet(s)
Infarctus du myocarde , Facteur de croissance endothéliale vasculaire de type A , Souris , Animaux , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Cellules cultivées , Différenciation cellulaire , Infarctus du myocarde/thérapie , Infarctus du myocarde/métabolisme , Myocytes cardiaques/métabolisme , Peptides/métabolisme , Cellules souches/métabolisme , Péricarde/métabolisme
19.
Int J Mol Sci ; 24(9)2023 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-37175796

RÉSUMÉ

New stem cell and extracellular-vesicle-based therapies have the potential to improve outcomes for the increasing number of patients with heart failure. Since neonates have a significantly enhanced regenerative ability, we hypothesized that extracellular vesicles isolated from Islet-1+ expressing neonatal human cardiovascular progenitors (CPCs) will induce transcriptomic changes associated with improved regenerative capability when co-cultured with CPCs derived from adult humans. In order to test this hypothesis, we isolated extracellular vesicles from human neonatal Islet-1+ CPCs, analyzed the extracellular vesicle content using RNAseq, and treated adult CPCs with extracellular vesicles derived from neonatal CPCs to assess their functional effect. AKT, ERBB, and YAP1 transcripts were elevated in adult CPCs treated with neonatal CPC-derived extracellular vesicles. YAP1 is lost after the neonatal period but can stimulate cardiac regeneration. Our results demonstrate that YAP1 and additional transcripts associated with improved cardiovascular regeneration, as well as the activation of the cell cycle, can be achieved by the treatment of adult CPCs with neonatal CPC-derived extracellular vesicles. Progenitor cells derived from neonates secrete extracellular vesicles with the potential to stimulate and potentially improve functional effects in adult CPCs used for cardiovascular repair.


Sujet(s)
Cellules souches adultes , Vésicules extracellulaires , Nouveau-né , Humains , Adulte , Myocytes cardiaques/métabolisme , Cellules cultivées , Cellules souches/métabolisme , Différenciation cellulaire
20.
Dev Cell ; 58(4): 257-266, 2023 02 27.
Article de Anglais | MEDLINE | ID: mdl-36809764

RÉSUMÉ

Organogenesis requires the orchestrated development of multiple cell lineages that converge, interact, and specialize to generate coherent functional structures, exemplified by transformation of the cardiac crescent into a four-chambered heart. Cardiomyocytes originate from the first and second heart fields, which make different regional contributions to the definitive heart. In this review, a series of recent single-cell transcriptomic analyses, together with genetic tracing experiments, are discussed, providing a detailed panorama of the cardiac progenitor cell landscape. These studies reveal that first heart field cells originate in a juxtacardiac field adjacent to extraembryonic mesoderm and contribute to the ventrolateral side of the cardiac primordium. In contrast, second heart field cells are deployed dorsomedially from a multilineage-primed progenitor population via arterial and venous pole pathways. Refining our knowledge of the origin and developmental trajectories of cells that build the heart is essential to address outstanding challenges in cardiac biology and disease.


Sujet(s)
Coeur , Myocytes cardiaques , Myocytes cardiaques/métabolisme , Lignage cellulaire/génétique , Mésoderme/métabolisme , Différenciation cellulaire/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE