Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 148(13)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34027990

RESUMO

Polycomb repressive complex 2 (PRC2) deposits H3K27me3 on chromatin to silence transcription. PRC2 broadly interacts with RNAs. Currently, the role of the RNA-PRC2 interaction in human cardiogenesis remains elusive. Here, we found that human-specific heart brake lncRNA 1 (HBL1) interacted with two PRC2 subunits, JARID2 and EED, in human pluripotent stem cells (hPSCs). Loss of JARID2, EED or HBL1 significantly enhanced cardiac differentiation from hPSCs. HBL1 depletion disrupted genome-wide PRC2 occupancy and H3K27me3 chromatin modification on essential cardiogenic genes, and broadly enhanced cardiogenic gene transcription in undifferentiated hPSCs and later-on differentiation. In addition, ChIP-seq revealed reduced EED occupancy on 62 overlapped cardiogenic genes in HBL1-/- and JARID2-/- hPSCs, indicating that the epigenetic state of cardiogenic genes was determined by HBL1 and JARID2 at pluripotency stage. Furthermore, after cardiac development occurs, the cytosolic and nuclear fractions of HBL1 could crosstalk via a conserved 'microRNA-1-JARID2' axis to modulate cardiogenic gene transcription. Overall, our findings delineate the indispensable role of HBL1 in guiding PRC2 function during early human cardiogenesis, and expand the mechanistic scope of lncRNA(s) that cytosolic and nuclear portions of HBL1 could coordinate to orchestrate human cardiogenesis.


Assuntos
Genoma , Organogênese , Células-Tronco Pluripotentes/metabolismo , Complexo Repressor Polycomb 2/genética , RNA Longo não Codificante/metabolismo , Diferenciação Celular , Cromatina , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Coração/crescimento & desenvolvimento , Histonas/genética , Humanos , MicroRNAs
2.
Stem Cells ; 41(4): 354-367, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-36715298

RESUMO

Mesendodermal specification and cardiac differentiation are key issues for developmental biology and heart regeneration medicine. Previously, we demonstrated that FAM122A, a highly conserved housekeeping gene, is an endogenous inhibitor of protein phosphatase 2A (PP2A) and participates in multifaceted physiological and pathological processes. However, the in vivo function of FAM122A is largely unknown. In this study, we observed that Fam122 deletion resulted in embryonic lethality with severe defects of cardiovascular developments and significantly attenuated cardiac functions in conditional cardiac-specific knockout mice. More importantly, Fam122a deficiency impaired mesendodermal specification and cardiac differentiation from mouse embryonic stem cells but showed no influence on pluripotent identity. Mechanical investigation revealed that the impaired differentiation potential was caused by the dysregulation of histone modification and Wnt and Hippo signaling pathways through modulation of PP2A activity. These findings suggest that FAM122A is a novel and critical regulator in mesendodermal specification and cardiac differentiation. This research not only significantly extends our understanding of the regulatory network of mesendodermal/cardiac differentiation but also proposes the potential significance of FAM122A in cardiac regeneration.


Assuntos
Células-Tronco Embrionárias , Processamento de Proteína Pós-Traducional , Animais , Camundongos , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo
3.
Stem Cells ; 41(12): 1142-1156, 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-37819786

RESUMO

In early embryogenesis, the primitive streak (PrS) generates the mesendoderm and is essential for organogenesis. However, because the PrS is a minute and transient tissue, elucidating the mechanism of its formation has been challenging. We performed comprehensive screening of 2 knockout mouse databases based on the fact that failure of PrS formation is lethal. We identified 812 genes involved in various cellular functions and responses that might be linked to PrS formation, with the category of greatest abundance being "Metabolism." In this study, we focused on genes of sphingolipid metabolism and investigated their roles in PrS formation using an in vitro mouse ES cell differentiation system. We show here that elevated intracellular ceramide negatively regulates gene expression essential for PrS formation and instead induces neurogenesis. In addition, sphingosine-1-phosphate (a ceramide derivative) positively regulates neural maturation. Our results indicate that ceramide regulates both PrS formation and the induction of neural differentiation.


Assuntos
Ceramidas , Linha Primitiva , Camundongos , Animais , Ceramidas/metabolismo , Linha Primitiva/metabolismo , Diferenciação Celular/genética , Neurogênese/genética , Fenótipo
4.
Stem Cells ; 41(2): 140-152, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36512477

RESUMO

The ability to differentiate human-induced pluripotent stem cells (hiPSCs) efficiently into defined cardiac lineages, such as cardiomyocytes and cardiac endothelial cells, is crucial to study human heart development and model cardiovascular diseases in vitro. The mechanisms underlying the specification of these cell types during human development are not well understood which limits fine-tuning and broader application of cardiac model systems. Here, we used the expression of ETV2, a master regulator of hematoendothelial specification in mice, to identify functionally distinct subpopulations during the co-differentiation of endothelial cells and cardiomyocytes from hiPSCs. Targeted analysis of single-cell RNA-sequencing data revealed differential ETV2 dynamics in the 2 lineages. A newly created fluorescent reporter line allowed us to identify early lineage-predisposed states and show that a transient ETV2-high-state initiates the specification of endothelial cells. We further demonstrated, unexpectedly, that functional cardiomyocytes can originate from progenitors expressing ETV2 at a low level. Our study thus sheds light on the in vitro differentiation dynamics of 2 important cardiac lineages.


Assuntos
Células Endoteliais , Células-Tronco Pluripotentes Induzidas , Animais , Camundongos , Humanos , Células Endoteliais/metabolismo , Miócitos Cardíacos/metabolismo , Regulação para Cima , Diferenciação Celular/genética , Endotélio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Differentiation ; 133: 77-87, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37506593

RESUMO

Precise spatiotemporal control of gene expression patterns is critical for normal development. Pluripotent stem cells, including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), with the ability of unlimited self-renewal and differentiation into any cell type, provide a unique tool for understanding the underlying mechanism of development and disease in a dish. N6-methyl-adenosine (m6A) modification is the most extensive internal mRNA modification, which regulates almost all aspects of mRNA metabolism and thus extensively participates in gene expression regulation. However, the role of m6A during cardiogenesis still needs to be fully elucidated. Here, we found that core components of m6A methyltransferase decreased during cardiomyocyte differentiation. Impeding m6A deposition, by either deleting the m6A methyltransferase Mettl3 or overexpressing m6A demethylase alkB homolog 5 (Alkbh5), at early stages of cardiac differentiation of mouse pluripotent stem cells, led to inhibition of cardiac gene activation and retardation of the outgrowth of embryoid bodies, whereas interfering m6A modification at later stages of differentiation had minimal effects. Consistently, stage-specific inhibition of METTL3 with METTL3 inhibitor STM2457 during human ESCs (hESCs) cardiac differentiation demonstrated a similarly pivotal role of METTL3 for the induction of mesodermal cells while dispensable function for later stages. In summary, our study reveals a stage-specific requirement of m6A on the cardiac differentiation of pluripotent stem cells and demonstrates that precise tuning of m6A level is critical for cardiac differentiation.


Assuntos
Metiltransferases , Células-Tronco Pluripotentes , Camundongos , Humanos , Animais , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Diferenciação Celular/genética , Células-Tronco Pluripotentes/metabolismo , RNA/metabolismo , RNA Mensageiro/genética
6.
Ecotoxicol Environ Saf ; 270: 115945, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38183750

RESUMO

Perfluorooctane sulfonate (PFOS), an endocrine-disrupting chemical pollutant, affects embryonic heart development; however, the mechanisms underlying its toxicity have not been fully elucidated. Here, Single-cell RNA sequencing (scRNA-seq) was used to investigate the overall effects of PFOS on myocardial differentiation from human embryonic stem cells (hESCs). Additionally, apoptosis, mitochondrial membrane potential, and ATP assays were performed. Downregulated cardiogenesis-related genes and inhibited cardiac differentiation were observed after PFOS exposure in vitro. The percentages of cardiomyocyte and cardiac progenitor cell clusters decreased significantly following exposure to PFOS, while the proportion of primitive endoderm cell was increased in PFOS group. Moreover, PFOS inhibited myocardial differentiation and blocked cellular development at the early- and middle-stage. A Gene Ontology analysis and pseudo-time trajectory illustrated that PFOS disturbed multiple processes related to cardiogenesis and oxidative phosphorylation in the mitochondria. Furthermore, PFOS decreased mitochondrial membrane potential and induced apoptosis. These results offer meaningful insights into the cardiogenic toxicity of PFOS exposure during heart formation as well as the adverse effects of PFOS on mitochondria.


Assuntos
Ácidos Alcanossulfônicos , Fluorocarbonos , Células-Tronco Embrionárias Humanas , Doenças Mitocondriais , Humanos , Fluorocarbonos/toxicidade , Fluorocarbonos/metabolismo , Miócitos Cardíacos , Análise de Sequência de RNA , Doenças Mitocondriais/metabolismo , Ácidos Alcanossulfônicos/toxicidade , Ácidos Alcanossulfônicos/metabolismo
7.
IUBMB Life ; 75(3): 196-206, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-34590780

RESUMO

The secretome of different cell types has been applied on in vitro and in vivo assays, indicating considerable therapeutic potential. However, the choice of the ideal cell type and culture conditions for obtaining the best set of soluble factors, as well as the assays to assess specific effects, remain subjects of vigorous debate. In this study, we used mass spectrometry to characterize the secretomes of ventricle derived-cardiac resident stromal cells (vCRSC) and human dermal fibroblasts (HDFs) and evaluate them in an effort to understand the niche specificity of biological responses toward different cellular behaviors, such as cell proliferation, adhesion, migration, and differentiation. It was interesting to note that the HDF and vCRSC secretomes were both able to induce proliferation and cardiac differentiation of H9c2 cells, as well as to increase the adhesion activity of H9c2 cells and human umbilical vein endothelial cells. Analysis of the secretome composition showed that the vCRSCs derived from different donors secreted a similar set of proteins. Despite the differences, almost half of the proteins identified in conditioned medium were common to both HDF and vCRSC. Consequently, a high number of common biological processes were identified in the secretomes of the two cell types, which could help to explain the similar results observed in the in vitro assays. We show that soluble factors secreted by both HDF and vCRSC are able to promote proliferation and differentiation of cardiomyoblasts in vitro. Our study indicates the possible use of vCRSC or HDF secretomes in acellular therapies for regenerative medicine.


Assuntos
Secretoma , Células Estromais , Humanos , Pele/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Fibroblastos/metabolismo
8.
Int J Mol Sci ; 24(3)2023 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-36768766

RESUMO

Cells of the cardiovascular system are physiologically exposed to a variety of mechanical forces fundamental for both cardiac development and functions. In this context, forces generated by actomyosin networks and those transmitted through focal adhesion (FA) complexes represent the key regulators of cellular behaviors in terms of cytoskeleton dynamism, cell adhesion, migration, differentiation, and tissue organization. In this study, we investigated the involvement of FAs on cardiomyocyte differentiation. In particular, vinculin and focal adhesion kinase (FAK) family, which are known to be involved in cardiac differentiation, were studied. Results revealed that differentiation conditions induce an upregulation of both FAK-Tyr397 and vinculin, resulting also in the translocation to the cell membrane. Moreover, the role of mechanical stress in contractile phenotype expression was investigated by applying a uniaxial mechanical stretching (5% substrate deformation, 1 Hz frequency). Morphological evaluation revealed that the cell shape showed a spindle shape and reoriented following the stretching direction. Substrate deformation resulted also in modification of the length and the number of vinculin-positive FAs. We can, therefore, suggest that mechanotransductive pathways, activated through FAs, are highly involved in cardiomyocyte differentiation, thus confirming their role during cytoskeleton rearrangement and cardiac myofilament maturation.


Assuntos
Adesões Focais , Adesões Focais/metabolismo , Vinculina/metabolismo , Adesão Celular/fisiologia , Membrana Celular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Diferenciação Celular
9.
Int J Mol Sci ; 24(15)2023 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-37569627

RESUMO

During cardiac differentiation, numerous factors contribute to the development of the heart. Understanding the molecular mechanisms underlying cardiac development will help combat cardiovascular disorders, among the leading causes of morbidity and mortality worldwide. Among the main mechanisms, we indeed find Cripto. Cripto is found in both the syncytiotrophoblast of ampullary pregnancies and the inner cell mass along the primitive streak as the second epithelial-mesenchymal transformation event occurs to form the mesoderm and the developing myocardium. At the same time, it is now known that cardiac signaling pathways are intimately intertwined with the expression of myomiRNAs, including miR-1. This miR-1 is one of the muscle-specific miRs; aberrant expression of miR-1 plays an essential role in cardiac diseases. Given this scenario, our study aimed to evaluate the inverse correlation between Cripto and miR-1 during heart development. We used in vitro models of the heart, represented by embryoid bodies (EBs) and embryonic carcinoma cell lines derived from an embryo-derived teratocarcinoma in mice (P19 cells), respectively. First, through a luciferase assay, we demonstrated that Cripto is a target of miR-1. Following this result, we observed that as the days of differentiation increased, the Cripto gene expression decreased, while the level of miR-1 increased; furthermore, after silencing miR-1 in P19 cells, there was an increase in Cripto expression. Moreover, inducing damage with a cobra cardiotoxin (CTX) in post-differentiation cells, we noted a decreased miR-1 expression and increased Cripto. Finally, in mouse cardiac biopsies, we observed by monitoring gene expression the distribution of Cripto and miR-1 in the right and left ventricles. These results allowed us to detect an inverse correlation between miR-1 and Cripto that could represent a new pharmacological target for identifying new therapies.


Assuntos
Fator de Crescimento Epidérmico , MicroRNAs , Animais , Camundongos , Diferenciação Celular , Fator de Crescimento Epidérmico/metabolismo , Coração , MicroRNAs/genética , MicroRNAs/metabolismo , Miocárdio/metabolismo
10.
Cell Commun Signal ; 20(1): 108, 2022 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-35850719

RESUMO

BACKGROUND: Macrophage phenotypes switch from proinflammatory (M1) to anti-inflammatory (M2) following myocardial injury. Implanted stem cells (e.g., induced pluripotent stem cells (iPSCs)) for cardiomyogenesis will inevitably contact the inflammatory environment at the myocardial infarction site. To understand how the macrophages affect the behavior of iPSCs, therefore, improve the therapeutic efficacy, we generated three macrophage subtypes and assessed their effects on the proliferation, cardiac differentiation, and maturation of iPSCs. METHODS: M0, M1, and M2 macrophages were polarized using cytokines, and their properties were confirmed by the expression of specific markers using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunofluorescence. The effects of macrophages on iPSCs were studied using Transwell co-culture models. The proliferative ability of iPSCs was investigated by cell counting and CCK-8 assays. The cardiac differentiation ability of iPSCs was determined by the cardiomyocyte (CM) yield. The maturation of CM was analyzed by the expression of cardiac-specific genes using RT-qPCR, the sarcomere organization using immunofluorescence, and the mitochondrial function using oxidative respiration analysis. RESULTS: The data showed that the co-culture of iPSCs with M0, M1, or M2 macrophages significantly decreased iPSCs' proliferative ability. M2 macrophages did not affect the CM yield during the cardiac differentiation of iPSCs. Still, they promoted the maturation of CM by improving sarcomeric structures, increasing contractile- and ion transport-associated gene expression, and enhancing mitochondrial respiration. M0 macrophages did not significantly affect the cardiomyogenesis ability of iPSCs during co-culture. In contrast, co-culture with M1 macrophages significantly reduced the cardiac differentiation and maturation of iPSCs. CONCLUSIONS: M1- or M2-polarized macrophages play critical roles in the proliferation, cardiac differentiation, and maturation of iPSCs, providing knowledge to improve the outcomes of stem cell regeneration therapy. Video abstract.


Assuntos
Células-Tronco Pluripotentes Induzidas , Diferenciação Celular , Proliferação de Células , Citocinas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Macrófagos/metabolismo
11.
Exp Cell Res ; 403(2): 112599, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33848551

RESUMO

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) create an unlimited cell source for basic and translational research. Depending on the maturity of cardiac cultures and the intended applications, obtaining hiPSC-CMs as a single-cell, monolayer or three-dimensional clusters can be challenging. Here, we defined strategies to replate hiPSC-CMs on early days (D15-30) or later more mature (D60-150) differentiation cultures. After generation of hiPSCs and derivation of cardiomyocytes, four dissociation reagents Collagenase A/B, Collagenase II, TrypLE, EDTA and five different extracellular matrix materials Laminin, iMatrix-511, Fibronectin, Matrigel, and Geltrex were comparatively evaluated by imaging, cell viability, and contraction analysis. For early cardiac differentiation cultures mimicking mostly the embryonic stage, the highest adhesion, cell viability, and beating frequencies were achieved by treatment with the TrypLE enzyme. Video-based contraction analysis demonstrated higher beating rates after replating compared to before treatment. For later differentiation days of more mature cardiac cultures, dissociation with EDTA and replating cells on Geltrex or Laminin-derivatives yielded better recovery. Cardiac clusters at various sizes were detected in several groups treated with collagenases. Collectively, our findings revealed the selection criteria of the dissociation approach and coating matrix for replating iPSC-CMs based on the maturity and the requirements of further downstream applications.


Assuntos
Técnicas de Cultura de Células , Meios de Cultura/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Adulto , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Reprogramação Celular/genética , Colágeno/farmacologia , Colagenases/farmacologia , Meios de Cultura/química , Combinação de Medicamentos , Feminino , Fibronectinas/farmacologia , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Insulina/análogos & derivados , Insulina/farmacologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Laminina/farmacologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Cultura Primária de Células , Proteoglicanas/farmacologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo
12.
Genomics ; 113(3): 1349-1365, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33713822

RESUMO

Yes-associated protein 1 (YAP1) is a transcriptional co-activator downstream of Hippo pathway. The pathway exerts crucial roles in organogenesis and its dysregulation is associated with the spreading of different cancer types. YAP1 gene encodes for multiple protein isoforms, whose specific functions are not well defined. We demonstrate the splicing of isoform-specific mRNAs is controlled in a stage- and tissue-specific fashion. We designed expression vectors encoding for the most-represented isoforms of YAP1 with either one or two WW domains and studied their specific signaling activities in YAP1 knock-out cell lines. YAP1 isoforms display both common and unique functions and activate distinct transcriptional programs, as the result of their unique protein interactomes. By generating TEAD-based transcriptional reporter cell lines, we demonstrate individual YAP1 isoforms display unique effects on cell proliferation and differentiation. Finally, we illustrate the complexity of the regulation of Hippo-YAP1 effector in physiological and in pathological conditions of the heart.


Assuntos
Proteínas de Ciclo Celular , Isoformas de RNA , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Sinalização YAP
13.
Int J Mol Sci ; 23(24)2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36555427

RESUMO

Human iPSC-derived self-organized cardiac tissues can be valuable for the development of platforms for disease modeling and drug screening, enhancing test accuracy and reducing pharmaceutical industry financial burden. However, current differentiation systems still rely on static culture conditions and specialized commercial microwells for aggregation, which hinders the full potential of hiPSC-derived cardiac tissues. Herein, we integrate cost-effective and reproducible manual aggregation of hiPSC-derived cardiac progenitors with Matrigel encapsulation and a dynamic culture to support hiPSC cardiac differentiation and self-organization. Manual aggregation at day 7 of cardiac differentiation resulted in 97% of beating aggregates with 78% of cTnT-positive cells. Matrigel encapsulation conjugated with a dynamic culture promoted cell migration and the creation of organized structures, with observed cell polarization and the creation of lumens. In addition, encapsulation increased buoyancy and decreased coalescence of the hiPSC-derived cardiac aggregates. Moreover, VEGF supplementation increased over two-fold the percentage of CD31-positive cells resulting in the emergence of microvessel-like structures. Thus, this study shows that the explored culture parameters support the self-organization of hiPSC-derived cardiac microtissues containing multiple cardiac cell types. Additional stimuli (e.g., BMP) in long-term scalable and fully automatized cultures can further potentiate highly structured and mature hiPSC-derived cardiac models, contributing to the development of reliable platforms for high-throughput drug screening and disease modeling.


Assuntos
Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Humanos , Miócitos Cardíacos/metabolismo , Células Cultivadas , Análise Custo-Benefício , Colágeno/metabolismo , Diferenciação Celular
14.
Int J Mol Sci ; 23(18)2022 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-36142249

RESUMO

Progesterone treatment is commonly employed to promote and support pregnancy. While maternal tissues are the main progesterone targets in humans and mice, its receptor (PGR) is expressed in the murine embryo, questioning its function during embryonic development. Progesterone has been previously associated with murine blastocyst development. Whether it contributes to lineage specification is largely unknown. Gastrulation initiates lineage specification and generation of the progenitors contributing to all organs. Cells passing through the primitive streak (PS) will give rise to the mesoderm and endoderm. Cells emerging posteriorly will form the extraembryonic mesodermal tissues supporting embryonic growth. Cells arising anteriorly will contribute to the embryonic heart in two sets of distinct progenitors, first (FHF) and second heart field (SHF). We found that PGR is expressed in a posterior-anterior gradient in the PS of gastrulating embryos. We established in vitro differentiation systems inducing posterior (extraembryonic) and anterior (cardiac) mesoderm to unravel PGR function. We discovered that PGR specifically modulates extraembryonic and cardiac mesoderm. Overexpression experiments revealed that PGR safeguards cardiac differentiation, blocking premature SHF progenitor specification and sustaining the FHF progenitor pool. This role of PGR in heart development indicates that progesterone administration should be closely monitored in potential early-pregnancy patients undergoing infertility treatment.


Assuntos
Gástrula , Gastrulação , Receptores de Progesterona , Animais , Diferenciação Celular , Feminino , Gástrula/fisiologia , Humanos , Mesoderma , Camundongos , Gravidez , Progesterona/metabolismo , Receptores de Progesterona/metabolismo
15.
Development ; 145(1)2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29217753

RESUMO

Several studies have demonstrated a multiphasic role for Wnt signaling during embryonic cardiogenesis and developed protocols that enrich for cardiac derivatives during in vitro differentiation of human pluripotent stem cells (hPSCs). However, few studies have investigated the role of Wnt signaling in the specification of cardiac progenitor cells (CPCs) toward downstream fates. Using transgenic mice and hPSCs, we tracked endothelial cells (ECs) that originated from CPCs expressing NKX2.5. Analysis of EC-fated CPCs at discrete phenotypic milestones during hPSC differentiation identified reduced Wnt activity as a hallmark of EC specification, and the enforced activation or inhibition of Wnt reduced or increased, respectively, the degree of vascular commitment within the CPC population during both hPSC differentiation and mouse embryogenesis. Wnt5a, which has been shown to exert an inhibitory influence on Wnt signaling during cardiac development, was dynamically expressed during vascular commitment of hPSC-derived CPCs, and ectopic Wnt5a promoted vascular specification of hPSC-derived and mouse embryonic CPCs.


Assuntos
Embrião de Mamíferos/metabolismo , Células Endoteliais/metabolismo , Coração/embriologia , Células-Tronco Pluripotentes/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Embrião de Mamíferos/citologia , Células Endoteliais/citologia , Humanos , Camundongos , Camundongos Transgênicos , Células-Tronco Pluripotentes/citologia , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
16.
Am J Physiol Regul Integr Comp Physiol ; 320(4): R547-R562, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33112656

RESUMO

Mitochondria play key roles in the differentiation and maturation of human cardiomyocytes (CMs). As human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) hold potential in the treatment of heart diseases, we sought to identify key mitochondrial pathways and regulators, which may provide targets for improving cardiac differentiation and maturation. Proteomic analysis was performed on enriched mitochondrial protein extracts isolated from hiPSC-CMs differentiated from dermal fibroblasts (dFCM) and cardiac fibroblasts (cFCM) at time points between 12 and 115 days of differentiation, and from adult and neonatal mouse hearts. Mitochondrial proteins with a twofold change at time points up to 120 days relative to 12 days were subjected to ingenuity pathway analysis (IPA). The highest upregulation was in metabolic pathways for fatty acid oxidation (FAO), the tricarboxylic acid (TCA) cycle, oxidative phosphorylation (OXPHOS), and branched chain amino acid (BCAA) degradation. The top upstream regulators predicted to be activated were peroxisome proliferator-activated receptor γ coactivator 1 α (PGC1-α), the insulin receptor (IR), and the retinoblastoma protein (Rb1) transcriptional repressor. IPA and immunoblotting showed upregulation of the mitochondrial LonP1 protease-a regulator of mitochondrial proteostasis, energetics, and metabolism. LonP1 knockdown increased FAO in neonatal rat ventricular cardiomyocytes (nRVMs). Our results support the notion that LonP1 upregulation negatively regulates FAO in cardiomyocytes to calibrate the flux between glucose and fatty acid oxidation. We discuss potential mechanisms by which IR, Rb1, and LonP1 regulate the metabolic shift from glycolysis to OXPHOS and FAO. These newly identified factors and pathways may help in optimizing the maturation of iPSC-CMs.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias Cardíacas/metabolismo , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/metabolismo , Biogênese de Organelas , Proteoma , Proteômica , Animais , Linhagem Celular , Linhagem da Célula , Metabolismo Energético , Humanos , Camundongos , Mitocôndrias Cardíacas/genética , Proteínas Mitocondriais/genética , Ratos , Fatores de Tempo
17.
Rev Cardiovasc Med ; 22(4): 1167-1171, 2021 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-34957760

RESUMO

Ischemic heart disease resulting from a myocardial infarction (MI), is a major health issue. Stem cell therapies may play an important role in this field. However, cardiomyocytes induced from stem cells are characterized by low rates of differentiation and immaturity. After transplantation into the damaged heart, they may even increase the risk of arrhythmias. Studies have demonstrated that electrical stimulation (ES) can promote the cardiac differentiation of stem cells. This review summarizes the latest research on the effects of applying different electrical stimulation (ES) parameters to different types of stem cells and the related mechanisms that may be involved.


Assuntos
Infarto do Miocárdio , Diferenciação Celular , Estimulação Elétrica , Humanos , Infarto do Miocárdio/terapia , Miócitos Cardíacos , Transplante de Células-Tronco/efeitos adversos , Células-Tronco
18.
Differentiation ; 115: 1-10, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32738735

RESUMO

The importance of oxygen tension in in vitro cultures and its effect on embryonic stem cell (ESC) differentiation has been widely acknowledged. Research has mainly focussed on ESC maintenance or on one line of differentiation and only few studies have examined the potential relation between oxygen tension during ESC maintenance and differentiation. In this study we investigated the influence of atmospheric (20%) versus physiologic (5%) oxygen tension in ESC cultures and their differentiation within the cardiac and neural embryonic stem cell tests (ESTc, ESTn). Oxygen tension was set at 5% or 20% and cells were kept in these conditions from starting up cell culture until use for differentiation. Under these oxygen tensions, ESC culture showed no differences in proliferation and gene and protein expression levels. Differentiation was either performed in the same or in the alternative oxygen tension compared to ESC culture creating four different experimental conditions. Cardiac differentiation in 5% instead of 20% oxygen resulted in reduced development of spontaneously beating cardiomyocytes and lower expression of cardiac markers Nkx2.5, Myh6 and MF20 (myosin), regardless whether ESC had been cultured in 5% or 20% oxygen tension. As compared to the control (20% oxygen during stem cell maintenance and differentiation), neural differentiation in 5% oxygen with ESC cultured in 20% oxygen led to more cardiac and neural crest cell differentiation. The opposite experimental condition of neural differentiation in 20% oxygen with ESC cultured in 5% oxygen resulted in more glial differentiation. ESC that were maintained and differentiated in 5% oxygen showed an increase in neural crest and oligodendrocytes as compared to 20% oxygen during stem cell maintenance and differentiation. This study showed major effects on ESC differentiation in ESTc and ESTn of oxygen tension, which is an important variable to consider when designing and developing a stem cell-based in vitro system.


Assuntos
Células-Tronco Embrionárias/metabolismo , Crista Neural/metabolismo , Células-Tronco Neurais/citologia , Organogênese/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Células-Tronco Embrionárias/citologia , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Crista Neural/citologia , Crista Neural/crescimento & desenvolvimento , Células-Tronco Neurais/metabolismo , Oxigênio/metabolismo
19.
Int J Mol Sci ; 22(19)2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34638648

RESUMO

Otic organoids have the potential to resolve current challenges in hearing loss research. The reproduction of the delicate and complex structure of the mammalian cochlea using organoids requires high efficiency and specificity. Recent attempts to strengthen otic organoids have focused on the effects of the Wnt signaling pathway on stem cell differentiation. One important aspect of this is the evaluation of undesirable effects of differentiation after Wnt activation. In the present study, we differentiated mouse embryonic stem cell embryoid bodies (EB) into otic organoids and observed two morphologies with different cell fates. EBs that underwent a core ejection process, or 'enucleation,' were similar to previously reported inner ear organoids. Meanwhile, EBs that retained their core demonstrated features characteristic of neural organoids. The application of a Wnt agonist during the maturation phase increased enucleation, as well as otic organoid formation, in turn leading to sensory hair cell-like cell generation. However, with a longer incubation period, Wnt activation also led to EBs with 'beating' organoids that exhibited spontaneous movement. This observation emphasizes the necessity of optimizing Wnt enhancement for the differentiation of specific cells, such as those found in the inner ear.


Assuntos
Diferenciação Celular/fisiologia , Cóclea/metabolismo , Cóclea/fisiologia , Organoides/metabolismo , Organoides/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Células Cultivadas , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/fisiologia , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/fisiologia , Células-Tronco Pluripotentes/metabolismo
20.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34209900

RESUMO

Adult human cardiomyocytes have an extremely limited proliferative capacity, which poses a great barrier to regenerative medicine and research. Human embryonic stem cells (hESCs) have been proposed as an alternative source to generate large numbers of clinical grade cardiomyocytes (CMs) that can have potential therapeutic applications to treat cardiac diseases. Previous studies have shown that bioactive lipids are involved in diverse cellular responses including cardiogenesis. In this study, we explored the novel function of the chemically synthesized bioactive lipid O-cyclic phytosphingosine-1-phosphate (cP1P) as an inducer of cardiac differentiation. Here, we identified cP1P as a novel factor that significantly enhances the differentiation potential of hESCs into cardiomyocytes. Treatment with cP1P augments the beating colony number and contracting area of CMs. Furthermore, we elucidated the molecular mechanism of cP1P regulating SMAD1/5/8 signaling via the ALK3/BMP receptor cascade during cardiac differentiation. Our result provides a new insight for cP1P usage to improve the quality of CM differentiation for regenerative therapies.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Esfingosina/análogos & derivados , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Lipídeos/química , Lipídeos/farmacologia , Miócitos Cardíacos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esfingosina/química , Esfingosina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA