Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 148(19)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34486669

RESUMO

Fibroblasts are activated to repair the heart following injury. Fibroblast activation in the mammalian heart leads to a permanent fibrotic scar that impairs cardiac function. In other organisms, such as zebrafish, cardiac injury is followed by transient fibrosis and scar-free regeneration. The mechanisms that drive scarring versus scar-free regeneration are not well understood. Here, we show that the homeobox-containing transcription factor Prrx1b is required for scar-free regeneration of the zebrafish heart as the loss of Prrx1b results in excessive fibrosis and impaired cardiomyocyte proliferation. Through lineage tracing and single-cell RNA sequencing, we find that Prrx1b is activated in epicardial-derived cells where it restricts TGFß ligand expression and collagen production. Furthermore, through combined in vitro experiments in human fetal epicardial-derived cells and in vivo rescue experiments in zebrafish, we conclude that Prrx1 stimulates Nrg1 expression and promotes cardiomyocyte proliferation. Collectively, these results indicate that Prrx1 is a key transcription factor that balances fibrosis and regeneration in the injured zebrafish heart. This article has an associated 'The people behind the papers' interview.


Assuntos
Proliferação de Células , Coração/fisiologia , Proteínas de Homeodomínio/metabolismo , Miócitos Cardíacos/metabolismo , Regeneração , Proteínas de Peixe-Zebra/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibrose , Proteínas de Homeodomínio/genética , Humanos , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Neuregulina-1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
2.
Proc Natl Acad Sci U S A ; 117(26): 15182-15192, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32554494

RESUMO

The anthracycline doxorubicin (Doxo) and its analogs daunorubicin (Daun), epirubicin (Epi), and idarubicin (Ida) have been cornerstones of anticancer therapy for nearly five decades. However, their clinical application is limited by severe side effects, especially dose-dependent irreversible cardiotoxicity. Other detrimental side effects of anthracyclines include therapy-related malignancies and infertility. It is unclear whether these side effects are coupled to the chemotherapeutic efficacy. Doxo, Daun, Epi, and Ida execute two cellular activities: DNA damage, causing double-strand breaks (DSBs) following poisoning of topoisomerase II (Topo II), and chromatin damage, mediated through histone eviction at selected sites in the genome. Here we report that anthracycline-induced cardiotoxicity requires the combination of both cellular activities. Topo II poisons with either one of the activities fail to induce cardiotoxicity in mice and human cardiac microtissues, as observed for aclarubicin (Acla) and etoposide (Etop). Further, we show that Doxo can be detoxified by chemically separating these two activities. Anthracycline variants that induce chromatin damage without causing DSBs maintain similar anticancer potency in cell lines, mice, and human acute myeloid leukemia patients, implying that chromatin damage constitutes a major cytotoxic mechanism of anthracyclines. With these anthracyclines abstained from cardiotoxicity and therapy-related tumors, we thus uncoupled the side effects from anticancer efficacy. These results suggest that anthracycline variants acting primarily via chromatin damage may allow prolonged treatment of cancer patients and will improve the quality of life of cancer survivors.


Assuntos
Antineoplásicos/efeitos adversos , Cromatina/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Doxorrubicina/efeitos adversos , Animais , Linhagem Celular , Doxorrubicina/análogos & derivados , Doxorrubicina/síntese química , Doxorrubicina/metabolismo , Doxorrubicina/uso terapêutico , Cardiopatias/induzido quimicamente , Histonas , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Camundongos
3.
Int J Mol Sci ; 22(9)2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-33946583

RESUMO

Molecular imaging of pathologic lesions can improve efficient detection of cancer and cardiovascular diseases. A shared pathophysiological feature is angiogenesis, the formation of new blood vessels. Endoglin (CD105) is a coreceptor for ligands of the Transforming Growth Factor-ß (TGF-ß) family and is highly expressed on angiogenic endothelial cells. Therefore, endoglin-based imaging has been explored to visualize lesions of the aforementioned diseases. This systematic review highlights the progress in endoglin-based imaging of cancer, atherosclerosis, myocardial infarction, and aortic aneurysm, focusing on positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), near-infrared fluorescence (NIRF) imaging, and ultrasound imaging. PubMed was searched combining the following subjects and their respective synonyms or relevant subterms: "Endoglin", "Imaging/Image-guided surgery". In total, 59 papers were found eligible to be included: 58 reporting about preclinical animal or in vitro models and one ex vivo study in human organs. In addition to exact data extraction of imaging modality type, tumor or cardiovascular disease model, and tracer (class), outcomes were described via a narrative synthesis. Collectively, the data identify endoglin as a suitable target for intraoperative and diagnostic imaging of the neovasculature in tumors, whereas for cardiovascular diseases, the evidence remains scarce but promising.


Assuntos
Doenças Cardiovasculares/diagnóstico por imagem , Endoglina/análise , Neoplasias/diagnóstico por imagem , Animais , Doenças Cardiovasculares/cirurgia , Humanos , Imageamento por Ressonância Magnética/métodos , Neoplasias/cirurgia , Imagem Óptica/métodos , Tomografia por Emissão de Pósitrons/métodos , Cirurgia Assistida por Computador/métodos , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Ultrassonografia/métodos
4.
Int J Mol Sci ; 22(4)2021 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-33670533

RESUMO

Hereditary hemorrhagic telangiectasia type 1 (HHT1) is a severe vascular disorder caused by mutations in the TGFß/BMP co-receptor endoglin. Endoglin haploinsufficiency results in vascular malformations and impaired neoangiogenesis. Furthermore, HHT1 patients display an impaired immune response. To date it is not fully understood how endoglin haploinsufficient immune cells contribute to HHT1 pathology. Therefore, we investigated the immune response during tissue repair in Eng+/- mice, a model for HHT1. Eng+/- mice exhibited prolonged infiltration of macrophages after experimentally induced myocardial infarction. Moreover, there was an increased number of inflammatory M1-like macrophages (Ly6Chigh/CD206-) at the expense of reparative M2-like macrophages (Ly6Clow/CD206+). Interestingly, HHT1 patients also showed an increased number of inflammatory macrophages. In vitro analysis revealed that TGFß-induced differentiation of Eng+/- monocytes into M2-like macrophages was blunted. Inhibiting BMP signaling by treating monocytes with LDN-193189 normalized their differentiation. Finally, LDN treatment improved heart function after MI and enhanced vascularization in both wild type and Eng+/- mice. The beneficial effect of LDN was also observed in the hind limb ischemia model. While blood flow recovery was hampered in vehicle-treated animals, LDN treatment improved tissue perfusion recovery in Eng+/- mice. In conclusion, BMPR kinase inhibition restored HHT1 macrophage imbalance in vitro and improved tissue repair after ischemic injury in Eng+/- mice.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Modelos Animais de Doenças , Endoglina/metabolismo , Infarto do Miocárdio/prevenção & controle , Pirazóis/farmacologia , Pirimidinas/farmacologia , Cicatrização/efeitos dos fármacos , Animais , Receptores de Proteínas Morfogenéticas Ósseas/genética , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Células Cultivadas , Endoglina/genética , Feminino , Heterozigoto , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Telangiectasia Hemorrágica Hereditária/genética , Telangiectasia Hemorrágica Hereditária/imunologia , Telangiectasia Hemorrágica Hereditária/metabolismo , Cicatrização/genética
5.
J Mol Cell Cardiol ; 143: 26-37, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32277975

RESUMO

RATIONALE: After cardiac damage, excessive neurite outgrowth (sympathetic hyperinnervation) can occur, which is related to ventricular arrhythmias/sudden cardiac death. Post-damage reactivation of epicardium causes epicardium-derived cells (EPDCs) to acquire a mesenchymal character, contributing to cardiac regeneration. Whether EPDCs also contribute to cardiac re/hyperinnervation, is unknown. AIM: To investigate whether mesenchymal EPDCs influence cardiac sympathetic innervation. METHODS AND RESULTS: Sympathetic ganglia were co-cultured with mesenchymal EPDCs and/or myocardium, and neurite outgrowth and sprouting density were assessed. Results showed a significant increase in neurite density and directional (i.e. towards myocardium) outgrowth when ganglia were co-cultured with a combination of EPDCs and myocardium, as compared to cultures with EPDCs or myocardium alone. In absence of myocardium, this outgrowth was not directional. Neurite differentiation of PC12 cells in conditioned medium confirmed these results via a paracrine effect, in accordance with expression of neurotrophic factors in myocardial explants co-cultured with EPDCs. Of interest, EPDCs increased the expression of nerve growth factor (NGF) in cultured, but not in fresh myocardium, possibly due to an "ischemic state" of cultured myocardium, supported by TUNEL and Hif1α expression. Cardiac tissues after myocardial infarction showed robust NGF expression in the infarcted, but not remote area. CONCLUSION: Neurite outgrowth and density increases significantly in the presence of EPDCs by a paracrine effect, indicating a new role for EPDCs in the occurrence of sympathetic re/hyperinnervation after cardiac damage.


Assuntos
Coração/inervação , Miocárdio/metabolismo , Pericárdio/metabolismo , Fibras Simpáticas Pós-Ganglionares/fisiologia , Animais , Apoptose/genética , Linhagem Celular Tumoral , Células Cultivadas , Gânglios Simpáticos/citologia , Gânglios Simpáticos/metabolismo , Humanos , Camundongos , Miocárdio/citologia , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/metabolismo , Crescimento Neuronal
6.
Pharmacol Res ; 127: 129-140, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28751220

RESUMO

Since the regenerative capacity of the adult mammalian heart is limited, cardiac injury leads to the formation of scar tissue and thereby increases the risk of developing compensatory heart failure. Stem cell therapy is a promising therapeutic approach but is facing problems with engraftment and clinical feasibility. Targeting an endogenous stem cell population could circumvent these limitations. The epicardium, a membranous layer covering the outside of the myocardium, is an accessible cell population which plays a key role in the developing heart. Epicardial cells undergo epithelial to mesenchymal transition (EMT), thus providing epicardial derived cells (EPDCs) that migrate into the myocardium and cooperate in myocardial vascularisation and compaction. In the adult heart, injury activates the epicardium, and an embryonic-like response is observed which includes EMT and differentiation of the EPDCs into cardiac cell types. Furthermore, paracrine communication between the epicardium and myocardium improves the regenerative response. The significant role of the epicardium has been shown in both the developing and the regenerating heart. Interestingly, the epicardial contribution to cardiac repair can be improved in several ways. In this review, an overview of the epicardial origin and fate will be given and potential therapeutic approaches will be discussed.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Pericárdio/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Animais , Insuficiência Cardíaca/terapia , Humanos , Transplante de Células-Tronco/métodos
7.
J Mol Cell Cardiol ; 90: 139-45, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26686990

RESUMO

The transcription factor Wilms' Tumor-1 (WT1) is essential for cardiac development. Deletion of Wt1 in mice results in disturbed epicardial and myocardial formation and lack of cardiac vasculature, causing embryonic lethality. Little is known about the role of WT1 in the human fetal heart. Therefore, as a first step, we analyzed the expression pattern of WT1 protein during human cardiac development from week 4 till week 20. WT1 expression was apparent in epicardial, endothelial and endocardial cells in a spatiotemporal manner. The expression of WT1 follows a pattern starting at the epicardium and extending towards the lumen of the heart, with differences in timing and expression levels between the atria and ventricles. The expression of WT1 in cardiac arterial endothelial cells reduces in time, whereas WT1 expression in the endothelial cells of cardiac veins and capillaries remains present at all stages studied. This study provides for the first time a detailed description of the expression of WT1 protein during human cardiac development, which indicates an important role for WT1 also in human cardiogenesis.


Assuntos
Células Endoteliais/metabolismo , Coração Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas WT1/genética , Endocárdio/crescimento & desenvolvimento , Endocárdio/metabolismo , Endocárdio/ultraestrutura , Células Endoteliais/ultraestrutura , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Endotélio Vascular/ultraestrutura , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/ultraestrutura , Feto , Humanos , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Pericárdio/crescimento & desenvolvimento , Pericárdio/metabolismo , Pericárdio/ultraestrutura , Proteínas WT1/metabolismo
8.
J Mol Cell Cardiol ; 89(Pt B): 251-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26527381

RESUMO

The cardiac autonomic nervous system (cANS) modulates heart rate, contraction force and conduction velocity. The embryonic chicken heart already responds to epinephrine prior to establishment of the cANS. The aim of this study was to define the regions of the heart that might participate in modulating the early autonomic response to epinephrine. Immunofluorescence analysis reveals expression of neural markers tubulin beta-3 chain and neural cell adhesion molecule in the epicardium during early development. In addition, expression of the ß2 adrenergic receptor, the receptor for epinephrine, was found in the epicardium. Ex-ovo micro-electrode recordings in hearts with inhibition of epicardial outgrowth showed a significantly reduced response of the heart rate to epinephrine compared to control hearts. This study suggests a role for the epicardium as autonomic modulator during early cardiac development.


Assuntos
Sistema Nervoso Autônomo/embriologia , Desenvolvimento Embrionário , Pericárdio/embriologia , Animais , Sistema Nervoso Autônomo/metabolismo , Biomarcadores/metabolismo , Embrião de Galinha , Epinefrina/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Neurônios/metabolismo , Pericárdio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Medula Espinal/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas WT1/metabolismo
9.
Stem Cell Reports ; 18(7): 1421-1435, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37390825

RESUMO

The epicardium plays an essential role in cardiogenesis by providing cardiac cell types and paracrine cues to the developing myocardium. The human adult epicardium is quiescent, but recapitulation of developmental features may contribute to adult cardiac repair. The cell fate of epicardial cells is proposed to be determined by the developmental persistence of specific subpopulations. Reports on this epicardial heterogeneity have been inconsistent, and data regarding the human developing epicardium are scarce. Here we specifically isolated human fetal epicardium and used single-cell RNA sequencing to define its composition and to identify regulators of developmental processes. Few specific subpopulations were observed, but a clear distinction between epithelial and mesenchymal cells was present, resulting in novel population-specific markers. Additionally, we identified CRIP1 as a previously unknown regulator involved in epicardial epithelial-to-mesenchymal transition. Overall, our human fetal epicardial cell-enriched dataset provides an excellent platform to study the developing epicardium in great detail.


Assuntos
Miocárdio , Pericárdio , Adulto , Humanos , Pericárdio/metabolismo , Miocárdio/metabolismo , Transição Epitelial-Mesenquimal , Feto/metabolismo , Análise de Célula Única , Proteínas de Transporte/metabolismo , Proteínas com Domínio LIM/metabolismo
10.
J Cell Mol Med ; 16(7): 1508-21, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22003890

RESUMO

We previously showed that human cardiomyocyte progenitor cells (hCMPCs) injected after myocardial infarction (MI) had differentiated into cardiomyocytes in vivo 3 months after MI. Here, we investigated the short-term (2 weeks) effects of hCMPCs on the infarcted mouse myocardium. MI was induced in immunocompromised (NOD/scid) mice, immediately followed by intramyocardial injection of hCMPCs labelled with enhanced green fluorescent protein (hCMPC group) or vehicle only (control group). Sham-operated mice served as reference. Cardiac performance was measured 2 and 14 days after MI by magnetic resonance imaging at 9.4 T. Left ventricular (LV) pressure-volume measurements were performed at day 15 followed by extensive immunohistological analysis. Animals injected with hCMPCs demonstrated a higher LV ejection fraction, lower LV end-systolic volume and smaller relaxation time constant than control animals 14 days after MI. hCMPCs engrafted in the infarcted myocardium, did not differentiate into cardiomyocytes, but increased vascular density and proliferation rate in the infarcted and border zone area of the hCMPC group. Injected hCMPCs engraft into murine infarcted myocardium where they improve LV systolic function and attenuate the ventricular remodelling process 2 weeks after MI. Since no cardiac differentiation of hCMPCs was evident after 2 weeks, the observed beneficial effects were most likely mediated by paracrine factors, targeting amongst others vascular homeostasis. These results demonstrate that hCMPCs can be applied to repair infarcted myocardium without the need to undergo differentiation into cardiomyocytes.


Assuntos
Diferenciação Celular , Isquemia/terapia , Mioblastos/transplante , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Animais , Células Cultivadas , Proteínas de Fluorescência Verde/metabolismo , Humanos , Isquemia/patologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mioblastos/citologia , Infarto do Miocárdio/patologia , Função Ventricular Esquerda , Remodelação Ventricular
11.
Front Cardiovasc Med ; 9: 758265, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36277772

RESUMO

A role for cardiac sympathetic hyperinnervation in arrhythmogenesis after myocardial infarction (MI) has increasingly been recognized. In humans and mice, the heart receives cervical as well as thoracic sympathetic contributions. In mice, superior cervical ganglia (SCG) have been shown to contribute significantly to myocardial sympathetic innervation of the left ventricular anterior wall. Of interest, the SCG is situated adjacent to the carotid body (CB), a small organ involved in oxygen and metabolic sensing. We investigated the remodeling of murine SCG and CB over time after MI. Murine SCG were isolated from control mice, as well as 24 h, 3 days, 7 days and 6 weeks after MI. SCG and CBs were stained for the autonomic nervous system markers ß3-tubulin, tyrosine hydroxylase (TH) and choline acetyltransferase (ChAT), as well as for the neurotrophic factors brain derived neurotropic factor (BDNF), nerve growth factor (NGF) and their tyrosine receptor kinase (pan TRK). Results show that after MI a significant increase in neuron size occurs, especially in the region bordering the CB. Co-expression of TH and ChAT is observed in SCG neuronal cells, but not in the CB. After MI, a significant decrease in ChAT intensity occurs, which negatively correlated with the increased cell size. In addition, an increase of BDNF and NGF at protein and mRNA levels was observed in both the CB and SCG. This upregulation of neurotropic factors coincides with the upregulation of their receptor within the SCG. These findings were concomitant with an increase in GAP43 expression in the SCG, which is known to contribute to axonal outgrowth and elongation. In conclusion, neuronal remodeling toward an increased adrenergic phenotype occurs in the SCG, which is possibly mediated by the CB and might contribute to pathological hyperinnervation after MI.

12.
J Cell Mol Med ; 15(12): 2723-34, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21306557

RESUMO

Previously we observed that cardiomyocyte progenitor cells (hCMPCs) isolated from the human heart differentiate spontaneously into cardiomyocytes and vascular cells when transplanted after myocardial infarction (MI) in the ischemic heart. After MI, deprivation of oxygen is the first major change in the cardiac environment. How cells handle hypoxia is highly cell type dependent. The effect of hypoxia on cardiac stem or progenitor cells remains to be elucidated. Here, we show for the first time that short- and long-term hypoxia have different effects on hCMPCs. Short-term hypoxia increased the migratory and invasive capacities of hCMPCs likely via mesenchymal transformation. Although long-term exposure to low oxygen levels did not induce differentiation of hCMPCs into mature cardiomyocytes or endothelial cells, it did increase their proliferation, stimulated the secretome of the cells which was shifted to a more anti-inflammatory profile and dampened the migration by altering matrix metalloproteinase (MMP) modulators. Interestingly, hypoxia greatly induced the expression of the extracellular matrix modulator thrombospondin-2 (TSP-2). Knockdown of TSP-2 resulted in increased proliferation, migration and MMP activity. In conclusion, short exposure to hypoxia increases migratory and invasive capacities of hCMPCs and prolonged exposure induces proliferation, an angiogenic secretion profile and dampens migration, likely controlled by TSP-2.


Assuntos
Hipóxia Celular , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Oxigênio/metabolismo , Células-Tronco/metabolismo , Western Blotting , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Ensaio de Imunoadsorção Enzimática , Feto/citologia , Feto/metabolismo , Regulação da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Metaloproteinases da Matriz/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Trombospondina 1/metabolismo , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo
13.
Front Cardiovasc Med ; 8: 750243, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34631842

RESUMO

The epicardium is an essential cell population during cardiac development. It contributes different cell types to the developing heart through epithelial-to-mesenchymal transition (EMT) and it secretes paracrine factors that support cardiac tissue formation. In the adult heart the epicardium is a quiescent layer of cells which can be reactivated upon ischemic injury, initiating an embryonic-like response in the epicardium that contributes to post-injury repair processes. Therefore, the epicardial layer is considered an interesting target population to stimulate endogenous repair mechanisms. To date it is still not clear whether there are distinct cell populations in the epicardium that contribute to specific lineages or aid in cardiac repair, or that the epicardium functions as a whole. To address this putative heterogeneity, novel techniques such as single cell RNA sequencing (scRNA seq) are being applied. In this review, we summarize the role of the epicardium during development and after injury and provide an overview of the most recent insights into the cellular composition and diversity of the epicardium.

14.
Front Cell Dev Biol ; 9: 765007, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34977017

RESUMO

The epicardium, the mesothelial layer covering the heart, is a crucial cell source for cardiac development and repair. It provides cells and biochemical signals to the heart to facilitate vascularization and myocardial growth. An essential element of epicardial behavior is epicardial epithelial to mesenchymal transition (epiMT), which is the initial step for epicardial cells to become motile and invade the myocardium. To identify targets to optimize epicardium-driven repair of the heart, it is vital to understand which pathways are involved in the regulation of epiMT. Therefore, we established a cell culture model for human primary adult and fetal epiMT, which allows for parallel testing of inhibitors and stimulants of specific pathways. Using this approach, we reveal Activin A and ALK4 signaling as novel regulators of epiMT, independent of the commonly accepted EMT inducer TGFß. Importantly, Activin A was able to induce epicardial invasion in cultured embryonic mouse hearts. Our results identify Activin A/ALK4 signaling as a modulator of epicardial plasticity which may be exploitable in cardiac regenerative medicine.

15.
Sci Transl Med ; 13(612): eabf2750, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34550725

RESUMO

Arrhythmogenic cardiomyopathy (ACM) is an inherited disorder often caused by pathogenic variants in desmosomal genes and characterized by progressive fibrotic and fat tissue accumulation in the heart. The cellular origin and responsible molecular mechanisms of fibro-fatty deposits have been a matter of debate, due to limitations in animal models recapitulating this phenotype. Here, we used human-induced pluripotent stem cell (hiPSC)­derived cardiac cultures, single-cell RNA sequencing (scRNA-seq), and explanted human ACM hearts to study the epicardial contribution to fibro-fatty remodeling in ACM. hiPSC-epicardial cells generated from patients with ACM showed spontaneous fibro-fatty cellular differentiation that was absent in isogenic controls. This was further corroborated upon siRNA-mediated targeting of desmosomal genes in hiPSC-epicardial cells generated from healthy donors. scRNA-seq analysis identified the transcription factor TFAP2A (activating enhancer-binding protein 2 alpha) as a key trigger promoting this process. Gain- and loss-of-function studies on hiPSC-epicardial cells and primary adult epicardial-derived cells demonstrated that TFAP2A mediated epicardial differentiation through enhancing epithelial-to-mesenchymal transition (EMT). Furthermore, examination of explanted hearts from patients with ACM revealed epicardial activation and expression of TFAP2A in the subepicardial mesenchyme. These data suggest that TFAP2A-mediated epicardial EMT underlies fibro-fatty remodeling in ACM, a process amenable to therapeutic intervention.


Assuntos
Displasia Arritmogênica Ventricular Direita , Diferenciação Celular , Humanos
16.
Cells ; 10(8)2021 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-34440833

RESUMO

RATIONALE: In recent decades, the great potential of human epicardium-derived cells (EPDCs) as an endogenous cell source for cardiac regeneration has been recognized. The limited availability and low proliferation capacity of primary human EPDCs and phenotypic differences between EPDCs obtained from different individuals hampers their reproducible use for experimental studies. AIM: To generate and characterize inducible proliferative adult human EPDCs for use in fundamental and applied research. METHODS AND RESULTS: Inducible proliferation of human EPDCs was achieved by doxycycline-controlled expression of simian virus 40 large T antigen (LT) with a repressor-based lentiviral Tet-On system. In the presence of doxycycline, these inducible EPDCs (iEPDCs) displayed high and long-term proliferation capacity. After doxycycline removal, LT expression ceased and the iEPDCs regained their cuboidal epithelial morphology. Similar to primary EPDCs, iEPDCs underwent an epithelial-to-mesenchymal transition (EMT) after stimulation with transforming growth factor ß3. This was confirmed by reverse transcription-quantitative polymerase chain reaction analysis of epithelial and mesenchymal marker gene expression and (immuno) cytochemical staining. Collagen gel-based cell invasion assays demonstrated that mesenchymal iEPDCs, like primary EPDCs, possess increased invasion and migration capacities as compared to their epithelial counterparts. Mesenchymal iEPDCs co-cultured with sympathetic ganglia stimulated neurite outgrowth similarly to primary EPDCs. CONCLUSION: Using an inducible LT expression system, inducible proliferative adult human EPDCs were generated displaying high proliferative capacity in the presence of doxycycline. These iEPDCs maintain essential epicardial characteristics with respect to morphology, EMT ability, and paracrine signaling following doxycycline removal. This renders iEPDCs a highly useful new in vitro model for studying human epicardial properties.


Assuntos
Pericárdio/metabolismo , Antígenos Transformantes de Poliomavirus/genética , Antígenos Transformantes de Poliomavirus/metabolismo , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Doxiciclina/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Gânglios Simpáticos/citologia , Gânglios Simpáticos/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Modelos Biológicos , Neuritos/fisiologia , Comunicação Parácrina/efeitos dos fármacos , Pericárdio/citologia , Fator de Crescimento Transformador beta3/farmacologia
17.
J Cell Mol Med ; 14(4): 861-70, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20219011

RESUMO

In the past years, cardiovascular progenitor cells have been isolated from the human heart and characterized. Up to date, no studies have been reported in which the developmental potential of foetal and adult cardiovascular progenitors was tested simultaneously. However, intrinsic differences will likely affect interpretations regarding progenitor cell potential and application for regenerative medicine. Here we report a direct comparison between human foetal and adult heart-derived cardiomyocyte progenitor cells (CMPCs). We show that foetal and adult CMPCs have distinct preferences to differentiate into mesodermal lineages. Under pro-angiogenic conditions, foetal CMPCs form more endothelial but less smooth muscle cells than adult CMPCs. Foetal CMPCs can also develop towards adipocytes, whereas neither foetal nor adult CMPCs show significant osteogenic differentiation. Interestingly, although both cell types differentiate into heart muscle cells, adult CMPCs give rise to electrophysiologically more mature cardiomyocytes than foetal CMPCs. Taken together, foetal CMPCs are suitable for molecular cell biology and developmental studies. The potential of adult CMPCs to form mature cardiomyocytes and smooth muscle cells may be essential for cardiac repair after transplantation into the injured heart.


Assuntos
Células-Tronco Adultas/citologia , Feto/citologia , Miócitos Cardíacos/citologia , Adipogenia , Adulto , Proliferação de Células , Humanos , Potenciais da Membrana/fisiologia , Neovascularização Fisiológica , Osteogênese
18.
Biochem Biophys Res Commun ; 392(3): 283-8, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20060813

RESUMO

Endoglin (ENG) promotes angiogenesis by enhancing activation of TGF-beta type I receptors ALK-1 and ALK-5. ALK-1 phosphorylates transcription factors SMAD1/5, which bind to BMP-responsive elements (BRE), whereas ALK-5 phosphorylates SMAD3, which binds to CAGA elements. Expression of ENG is increased during myocardial infarction (MI). We investigated which ENG signaling pathway is activated in endothelial cells during hypoxia. Expression of ENG, ALK-1, ALK-5, and phosphorylated SMAD1/3/5 by immunostaining and immunoblotting in a mouse model of myocardial infarction (MI) and in hypoxic human aortic endothelial cells (HAECs) was evaluated. Activation of BRE and CAGA was measured by luciferase assays in cells transfected with plasmids expressing ENG or ALK-1 and the number of cells was quantified. mRNA expression of the target genes of TGF-beta signaling, ID1 and BCL-X, was quantified by real-time RT-PCR. Expression of ENG, ALK-1 and phosphorylated SMAD1/5, but not ALK-5 or phosphorylated SMAD3, was significantly increased in hypoxic endothelial cells in vivo and in vitro. Overexpression of both ENG and ALK-1 significantly increased BRE but not CAGA activity, expression of ID1 and BCL-X and the number of HAECs at hypoxia. ENG/ALK-1 signaling is one of the factors that regulate endothelial cell activity during adaptive cardiac angiogenesis.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Antígenos CD/metabolismo , Endotélio Vascular/metabolismo , Infarto do Miocárdio/metabolismo , Receptores de Superfície Celular/metabolismo , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Animais , Hipóxia Celular , Endoglina , Regulação da Expressão Gênica , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Camundongos , Infarto do Miocárdio/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Elementos de Resposta , Proteína Smad3/metabolismo , Proteína bcl-X/genética
19.
Curr Mol Med ; 9(3): 287-98, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19355911

RESUMO

Cardiovascular disease is one of the leading causes of morbidity and mortality around the world. Even after successful revascularization in coronary artery disease, cell death continues and the loss of cardiomyocytes eventually leads to progressive ventricular dilation and heart dysfunction. The notion of repairing or regenerating lost myocardium via cell-based therapies remains highly appealing. The recent identification of human stem cells, including embryonic stem cells and adult stem cells, has raised optimism for the development of a new therapy. This new cell-therapy and the concept of regenerative medicine is aimed at restoring the damaged myocardium, both vasculature and muscle. Here, we review the stem cell field and other available cell sources for myocardial regeneration, focusing on the up-to-date status of stem cell biology, recent laboratory advances and the current clinical applications. In addition, the limitations and practical hurdles that need urgent solution before more extensive applications become feasible are also discussed.


Assuntos
Doenças Cardiovasculares/terapia , Terapia Baseada em Transplante de Células e Tecidos , Miocárdio , Regeneração/fisiologia , Adipócitos Marrons/fisiologia , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Doenças Cardiovasculares/patologia , Ensaios Clínicos como Assunto , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Humanos , Mioblastos/citologia , Mioblastos/fisiologia , Miocárdio/citologia , Miocárdio/metabolismo , Medicina Regenerativa/métodos , Transplante de Células-Tronco
20.
Biomolecules ; 10(3)2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32150964

RESUMO

The epicardium, the outer layer of the heart, has been of interest in cardiac research due to its vital role in the developing and diseased heart. During development, epicardial cells are active and supply cells and paracrine cues to the myocardium. In the injured adult heart, the epicardium is re-activated and recapitulates embryonic behavior that is essential for a proper repair response. Two indispensable processes for epicardial contribution to heart tissue formation are epithelial to mesenchymal transition (EMT), and tissue invasion. One of the key groups of cytokines regulating both EMT and invasion is the transforming growth factor ß (TGFß) family, including TGFß and Bone Morphogenetic Protein (BMP). Abundant research has been performed to understand the role of TGFß family signaling in the developing epicardium. However, less is known about signaling in the adult epicardium. This review provides an overview of the current knowledge on the role of TGFß in epicardial behavior both in the development and in the repair of the heart. We aim to describe the presence of involved ligands and receptors to establish if and when signaling can occur. Finally, we discuss potential targets to improve the epicardial contribution to cardiac repair as a starting point for future investigation.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Transição Epitelial-Mesenquimal , Pericárdio/fisiologia , Regeneração , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA