Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Dis Model Mech ; 16(5)2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36263604

RESUMO

There is an unmet need for treatments that prevent the progressive cardiac dysfunction following myocardial infarction. Mesenchymal stem/stromal cells (MSCs) are under investigation for cardiac repair; however, culture expansion prior to transplantation is hindering their homing and reparative abilities. Pharmacological mobilisation could be an alternative to MSC transplantation. Here, we report that endogenous MSCs mobilise into the circulation at day 5 post myocardial infarction in male Lewis rats. This mobilisation can be significantly increased by using a combination of the FDA-approved drugs mirabegron (ß3-adrenoceptor agonist) and AMD3100 (CXCR4 antagonist). Blinded cardiac magnetic resonance imaging analysis showed the treated group to have increased left ventricular ejection fraction and decreased end systolic volume at 5 weeks post myocardial infarction. The mobilised group had a significant decrease in plasma IL-6 and TNF-α levels, a decrease in interstitial fibrosis, and an increase in the border zone blood vessel density. Conditioned medium from blood-derived MSCs supported angiogenesis in vitro, as shown by tube formation and wound healing assays. Our data suggest a novel pharmacological strategy that enhances myocardial infarction-induced MSC mobilisation and improves cardiac function after myocardial infarction.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Infarto do Miocárdio , Ratos , Animais , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Volume Sistólico , Função Ventricular Esquerda , Ratos Endogâmicos Lew , Infarto do Miocárdio/patologia
2.
ESC Heart Fail ; 9(1): 224-235, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34931757

RESUMO

AIMS: Hippo signalling is an evolutionarily conserved pathway that controls organ size by regulating apoptosis, cell proliferation, and stem cell self-renewal. Recently, the pathway has been shown to exert powerful growth regulatory activity in cardiomyocytes. However, the functional role of this stress-related and cell death-related pathway in the human heart and cardiomyocytes is not known. In this study, we investigated the role of the transcriptional co-activators of Hippo signalling, YAP and TAZ, in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in response to cardiotoxic agents and investigated the effects of modulating the pathway on cardiomyocyte function and survival. METHODS AND RESULTS: RNA-sequencing analysis of human heart samples with doxorubicin-induced end-stage heart failure and healthy controls showed that YAP and ERBB2 (HER2) as upstream regulators of differentially expressed genes correlated with doxorubicin treatment. Thus, we tested the effects of doxorubicin on hiPSC-CMs in vitro. Using an automated high-content screen of 96 clinically relevant antineoplastic and cardiotherapeutic drugs, we showed that doxorubicin induced the highest activation of YAP/TAZ nuclear translocation in both hiPSC-CMs and control MCF7 breast cancer cells. The overexpression of YAP rescued doxorubicin-induced cell loss in hiPSC-CMs by inhibiting apoptosis and inducing proliferation. In contrast, silencing of YAP and TAZ by siRNAs resulted in elevated mitochondrial membrane potential loss in response to doxorubicin. hiPSC-CM calcium transients did not change in response to YAP/TAZ silencing. CONCLUSIONS: Our results suggest that Hippo signalling is involved in clinical anthracycline-induced cardiomyopathy. Modelling with hiPSC-CMs in vitro showed similar responses to doxorubicin as adult cardiomyocytes and revealed a potential cardioprotective effect of YAP in doxorubicin-induced cardiotoxicity.


Assuntos
Cardiomiopatias , Fatores de Transcrição , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/metabolismo , Cardiotoxicidade/etiologia , Doxorrubicina/efeitos adversos , Doxorrubicina/metabolismo , Humanos , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/farmacologia , Proteínas de Sinalização YAP
4.
ACS Appl Mater Interfaces ; 13(28): 32624-32639, 2021 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-34228435

RESUMO

Organ dysfunction is a major cause of morbidity and mortality. Transplantation is typically the only definitive cure, challenged by the lack of sufficient donor organs. Tissue engineering encompasses the development of biomaterial scaffolds to support cell attachment, proliferation, and differentiation, leading to tissue regeneration. For efficient clinical translation, the forming technology utilized must be suitable for mass production. Herein, uniaxial polyhydroxyalkanoate scaffolds manufactured by pressurized gyration, a hybrid scalable spinning technique, are successfully used in bone, nerve, and cardiovascular applications. Chorioallantoic membrane and in vivo studies provided evidence of vascularization, collagen deposition, and cellular invasion for bone tissue engineering. Highly efficient axonal outgrowth was observed in dorsal root ganglion-based 3D ex vivo models. Human induced pluripotent stem cell derived cardiomyocytes exhibited a mature cardiomyocyte phenotype with optimal calcium handling. This study confirms that engineered polyhydroxyalkanoate-based gyrospun fibers provide an exciting and unique toolbox for the development of scalable scaffolds for both hard and soft tissue regeneration.


Assuntos
Células/metabolismo , Poli-Hidroxialcanoatos/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Galinhas , Módulo de Elasticidade , Gânglios Espinais/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Porosidade , Pressão , Ratos , Rotação , Células de Schwann/metabolismo
5.
Mol Ther Methods Clin Dev ; 20: 39-53, 2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33335946

RESUMO

During normal- and patho-physiological situations, the behavior of the beta2-adrenoreceptor (ß2AR) is influenced by polymorphic variants. The functional impact of such polymorphisms has been suggested from data derived from genetic association studies, in vitro experiments with primary cells, and transgenic overexpression models. However, heterogeneous genetic background and non-physiological transgene expression levels confound interpretation, leading to conflicting mechanistic conclusions. To overcome these limitations, we used CRISPR/Cas9 gene editing technology in human pluripotent stem cells (hPSCs) to create a unique suite of four isogenic homozygous variants at amino acid positions 16(G/R) and 27(G/Q), which reside in the N terminus of the ß2AR. By producing cardiomyocytes from these hPSC lines, we determined that at a functional level ß2AR signaling dominated over ß1AR . Examining changes in beat rates and responses to isoprenaline, Gi coupling, cyclic AMP (cAMP) production, downregulation, and desensitization indicated that responses were often heightened for the GE variant, implying differential dominance of both polymorphic location and amino acid substitution. This finding was corroborated, since GE showed hypersensitivity to doxorubicin-induced cardiotoxicity relative to GQ and RQ variants. Thus, understanding the effect of ß2AR polymorphisms on cardiac response to anticancer therapy may provide a route for personalized medicine and facilitate immediate clinical impact.

6.
Cells ; 9(10)2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-33053822

RESUMO

The ability to differentiate induced-pluripotent stem cells into cardiomyocytes (iPSC-CMs) has opened up novel avenues for potential cardiac therapies. However, iPSC-CMs exhibit a range of somewhat immature functional properties. This study explored the development of the beta-adrenergic receptor (ßAR) pathway, which is crucial in regulating contraction and signifying the health and maturity of myocytes. We explored the compartmentation of ß2AR-signalling and phosphodiesterases (PDEs) in caveolae, as functional nanodomains supporting the mature phenotype. Förster Resonance Energy Transfer (FRET) microscopy was used to study the cyclic adenosine monophosphate (cAMP) levels in iPSC-CMs at day 30, 60, and 90 following ßAR subtype-specific stimulation. Subsequently, the PDE2, PDE3, and PDE4 activity was investigated using specific inhibitors. Cells were treated with methyl-ß-cyclodextrin (MßCD) to remove cholesterol as a method of decompartmentalising ß2AR. As iPSC-CMs mature with a prolonged culture time, the caveolae density is increased, leading to a reduction in the overall cytoplasmic cAMP signal stimulated through ß2AR (but not ß1AR). Pan-phosphodiesterase inhibition or caveolae depletion leads to an increase in the ß2AR-stimulated cytoplasmic cAMP. Moreover, with time in culture, the increase in the ßAR-dependent cytoplasmic cAMP becomes more sensitive to cholesterol removal. The regulation of the ß2AR response by PDE2 and 4 is similarly increased with the time in culture. We conclude that both the ß2AR and PDEs are restricted to the caveolae nanodomains, and thereby exhibit a tighter spatial restriction over the cAMP signal in late-stage compared to early iPSC-CMs.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Cavéolas/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Insuficiência Cardíaca/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/fisiologia , Diester Fosfórico Hidrolases/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais
7.
Sci Rep ; 10(1): 13016, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32747668

RESUMO

Ischemic heart disease remains the foremost cause of death globally, with survivors at risk for subsequent heart failure. Paradoxically, cell therapies to offset cardiomyocyte loss after ischemic injury improve long-term cardiac function despite a lack of durable engraftment. An evolving consensus, inferred preponderantly from non-human models, is that transplanted cells benefit the heart via early paracrine signals. Here, we tested the impact of paracrine signals on human cardiomyocytes, using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) as the target of mouse and human cardiac mesenchymal stromal cells (cMSC) with progenitor-like features. In co-culture and conditioned medium studies, cMSCs markedly inhibited human cardiomyocyte death. Little or no protection was conferred by mouse tail tip or human skin fibroblasts. Consistent with the results of transcriptomic profiling, functional analyses showed that the cMSC secretome suppressed apoptosis and preserved cardiac mitochondrial transmembrane potential. Protection was independent of exosomes under the conditions tested. In mice, injecting cMSC-conditioned media into the infarct border zone reduced apoptotic cardiomyocytes > 70% locally. Thus, hPSC-CMs provide an auspicious, relevant human platform to investigate extracellular signals for cardiac muscle survival, substantiating human cardioprotection by cMSCs, and suggesting the cMSC secretome or its components as potential cell-free therapeutic products.


Assuntos
Células-Tronco Mesenquimais/citologia , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Células Estromais/citologia , Animais , Técnicas de Cocultura , Meios de Cultivo Condicionados , Humanos , Camundongos
8.
Sci Rep ; 10(1): 12060, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32694738

RESUMO

Given the poor track record to date of animal models for creating cardioprotective drugs, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have been proposed as a therapeutically relevant human platform to guide target validation and cardiac drug development. Mitogen-Activated Protein Kinase Kinase Kinase Kinase-4 (MAP4K4) is an "upstream" member of the MAPK superfamily that is implicated in human cardiac muscle cell death from oxidative stress, based on gene silencing and pharmacological inhibition in hPSC-CMs. A further role for MAP4K4 was proposed in heart muscle cell death triggered by cardiotoxic anti-cancer drugs, given its reported activation in failing human hearts with doxorubicin (DOX) cardiomyopathy, and its activation acutely by DOX in cultured cardiomyocytes. Here, we report successful protection from DOX in two independent hPSC-CM lines, using two potent, highly selective MAP4K4 inhibitors. The MAP4K4 inhibitors enhanced viability and reduced apoptosis at otherwise lethal concentrations of DOX, and preserved cardiomyocyte function, as measured by spontaneous calcium transients, at sub-maximal ones. Notably, in contrast, no intereference was seen in tumor cell killing, caspase activation, or mitochondrial membrane dissipation by DOX, in human cancer cell lines. Thus, MAP4K4 is a plausible, tractable, selective therapeutic target in DOX-induced human heart muscle cell death.


Assuntos
Cardiotônicos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Antraciclinas/efeitos adversos , Antineoplásicos/efeitos adversos , Apoptose/efeitos dos fármacos , Cálcio , Cardiotoxicidade/etiologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Doxorrubicina/farmacologia , Humanos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
9.
Cell Rep ; 23(2): 459-469, 2018 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-29642004

RESUMO

Cardiomyocytes from the apex but not the base of the heart increase their contractility in response to ß2-adrenoceptor (ß2AR) stimulation, which may underlie the development of Takotsubo cardiomyopathy. However, both cell types produce comparable cytosolic amounts of the second messenger cAMP. We investigated this discrepancy using nanoscale imaging techniques and found that, structurally, basal cardiomyocytes have more organized membranes (higher T-tubular and caveolar densities). Local membrane microdomain responses measured in isolated basal cardiomyocytes or in whole hearts revealed significantly smaller and more short-lived ß2AR/cAMP signals. Inhibition of PDE4, caveolar disruption by removing cholesterol or genetic deletion of Cav3 eliminated differences in local cAMP production and equilibrated the contractile response to ß2AR. We conclude that basal cells possess tighter control of cAMP because of a higher degree of signaling microdomain organization. This provides varying levels of nanostructural control for cAMP-mediated functional effects that orchestrate macroscopic, regional physiological differences within the heart.


Assuntos
Membrana Celular/química , AMP Cíclico/metabolismo , Coração/anatomia & histologia , Receptores Adrenérgicos beta 2/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Caveolina 3/deficiência , Caveolina 3/genética , Membrana Celular/metabolismo , Colesterol/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Feminino , Coração/fisiologia , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Knockout , Contração Muscular/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/genética , Transdução de Sinais/efeitos dos fármacos , beta-Ciclodextrinas/farmacologia
10.
Pflugers Arch ; 470(3): 559-570, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29297096

RESUMO

Estrogen and ß2-adrenergic receptors (ß2AR) play important roles in the processes that protect the heart. Here, we investigated how ovariectomy influenced the ß2AR downstream pathways in the context of catecholaminergic stress. In vivo and in vitro stress models were developed in female Sprague-Dawley (SD) rats by epinephrine (Epi) treatments. The cardiac function was evaluated at in vivo and in vitro levels in terms of contraction, rhythm, and injury. We found that myocardial contractility was not significantly different between Sham and ovariectomized (OVX) group rats in the normal state. However, Epi pretreatment decreased the contractility and increased abnormal rhythms especially in OVX group, which were attributed to lack of estrogen. Inhibition of the ß2AR-Gi-PI3K/p38MAPK pathway with ICI118,551, PTX or LY294002 increased contractility and aggravated Epi-induced injury on cardiomyocytes, decreased p38MAPK phosphorylation, and only increased arrhythmia in Sham group. These results indicated that OVX exacerbated cardiac injury and abnormal rhythms through ß2AR-Gi-PI3K and ß2AR-Gi-p38MAPK pathways, respectively. In normal state, the levels of activated Gi were similar in both groups, but those of cAMP and activated Gs were higher in OVX group. Epi treatment increased activated Gi (especially in Sham group) and activated Gs and cAMP in Sham group but decreased it in OVX group. These results suggested that estrogen increased the Gi activity in normal and stress states and Gs activity in stress state. These results indicated that lack of estrogen impaired the ß2AR-Gs/Gi coupling during stress which compromised cardiac contractility and increased abnormal rhythms.


Assuntos
Arritmias Cardíacas/metabolismo , Estrogênios/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Estresse Psicológico/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Arritmias Cardíacas/etiologia , Células Cultivadas , Epinefrina/farmacologia , Feminino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/complicações , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
J Tissue Eng Regen Med ; 12(1): e495-e512, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-27689781

RESUMO

Cardiac tissue engineering (CTE) is currently a prime focus of research because of an enormous clinical need. In the present work, a novel functional material, poly(3-hydroxyoctanoate), P(3HO), a medium chain-length polyhydroxyalkanoate (PHA), produced using bacterial fermentation, was studied as a new potential material for CTE. Engineered constructs with improved mechanical properties, crucial for supporting the organ during new tissue regeneration, and enhanced surface topography, to allow efficient cell adhesion and proliferation, were fabricated. Results showed that the mechanical properties of the final patches were close to that of cardiac muscle. Biocompatibility of neat P(3HO) patches, assessed using neonatal ventricular rat myocytes (NVRM), showed that the polymer was as good as collagen in terms of cell viability, proliferation and adhesion. Enhanced cell adhesion and proliferation properties were observed when porous and fibrous structures were incorporated into the patches. In addition, no deleterious effect was observed on adult cardiomyocyte contraction when cardiomyocytes were seeded on the P(3HO) patches. Hence, P(3HO)-based multifunctional cardiac patches are promising constructs for efficient CTE. This work will have a positive impact on the development of P(3HO) and other PHAs as a novel new family of biodegradable functional materials with huge potential in a range of different biomedical applications, particularly CTE, leading to further interest and exploitation of these materials. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Caprilatos/farmacologia , Coração/fisiologia , Teste de Materiais , Engenharia Tecidual/métodos , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Coração/efeitos dos fármacos , Camundongos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Temperatura , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Stem Cell Reports ; 9(5): 1415-1422, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-28988988

RESUMO

Tissue engineering offers an exciting possibility for cardiac repair post myocardial infarction. We assessed the effects of combined polyethylene glycol hydrogel (PEG), human induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM), and erythropoietin (EPO) therapy in a rat model of myocardial infarction. PEG with/out iPSC-CMs and EPO; iPSC-CMs in saline; or saline alone was injected into infarcted hearts shortly after infarction. Injection of almost any combination of the therapeutics limited acute elevations in chamber volumes. After 10 weeks, attenuation of ventricular remodeling was identified in all groups that received PEG injections, while ejection fractions were significantly increased in the gel-EPO, cell, and gel-cell-EPO groups. In all treatment groups, infarct thickness was increased and regions of muscle were identified within the scar. However, no grafted cells were detected. Hence, iPSC-CM-encapsulating bioactive hydrogel therapy can improve cardiac function post myocardial infarction and increase infarct thickness and muscle content despite a lack of sustained donor-cell engraftment.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Animais , Células Cultivadas , Eritropoetina/administração & dosagem , Eritropoetina/uso terapêutico , Humanos , Hidrogéis/química , Células-Tronco Pluripotentes Induzidas/citologia , Injeções Intralesionais , Masculino , Miócitos Cardíacos/citologia , Polietilenoglicóis/química , Ratos , Ratos Nus
13.
Stem Cell Res Ther ; 8(1): 208, 2017 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-28962641

RESUMO

BACKGROUND: Human dental mesenchymal stem cells (MSCs) are considered as highly accessible and attractive MSCs for use in regenerative medicine, yet some of their features are not as well characterized as other MSCs. Hypoxia-preconditioning and hypoxia-inducible factor 1 (HIF-1) alpha overexpression significantly improves MSC therapeutics, but the mechanisms involved are not fully understood. In the present study, we characterize immunomodulatory properties of dental MSCs and determine changes in their ability to modulate adaptive and innate immune populations after HIF-1 alpha overexpression. METHODS: Human dental MSCs were stably transduced with green fluorescent protein (GFP-MSCs) or GFP-HIF-1 alpha lentivirus vectors (HIF-MSCs). A hypoxic-like metabolic profile was confirmed by mitochondrial and glycolysis stress test. Capacity of HIF-MSCs to modulate T-cell activation, dendritic cell differentiation, monocyte migration, and polarizations towards macrophages and natural killer (NK) cell lytic activity was assessed by a number of functional assays in co-cultures. The expression of relevant factors were determined by polymerase chain reaction (PCR) analysis and enzyme-linked immunosorbent assay (ELISA). RESULTS: While HIF-1 alpha overexpression did not modify the inhibition of T-cell activation by MSCs, HIF-MSCs impaired dendritic cell differentiation more efficiently. In addition, HIF-MSCs showed a tendency to induce higher attraction of monocytes, which differentiate into suppressor macrophages, and exhibited enhanced resistance to NK cell-mediated lysis, which supports the improved therapeutic capacity of HIF-MSCs. HIF-MSCs also displayed a pro-angiogenic profile characterized by increased expression of CXCL12/SDF1 and CCL5/RANTES and complete loss of CXCL10/IP10 transcription. CONCLUSIONS: Immunomodulation and expression of trophic factors by dental MSCs make them perfect candidates for cell therapy. Overexpression of HIF-1 alpha enhances these features and increases their resistance to allogenic NK cell lysis and, hence, their potential in vivo lifespan. Our results further support the use of HIF-1 alpha-expressing dental MSCs for cell therapy in tissue injury and immune disorders.


Assuntos
Polpa Dentária/citologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imunomodulação , Células-Tronco Mesenquimais/imunologia , Diferenciação Celular , Células Cultivadas , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/imunologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Células-Tronco Mesenquimais/citologia
14.
Cardiovasc Res ; 113(7): 770-782, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28505272

RESUMO

AIMS: Cardiomyocyte ß2-adrenergic receptor (ß2AR) cyclic adenosine monophosphate (cAMP) signalling is regulated by the receptors' subcellular location within transverse tubules (T-tubules), via interaction with structural and regulatory proteins, which form a signalosome. In chronic heart failure (HF), ß2ARs redistribute from T-tubules to the cell surface, which disrupts functional signalosomes and leads to diffuse cAMP signalling. However, the functional consequences of structural changes upon ß2AR-cAMP signalling during progression from hypertrophy to advanced HF are unknown. METHODS AND RESULTS: Rat left ventricular myocytes were isolated at 4-, 8-, and 16-week post-myocardial infarction (MI), ß2ARs were stimulated either via whole-cell perfusion or locally through the nanopipette of the scanning ion conductance microscope. cAMP release was measured via a Förster Resonance Energy Transfer-based sensor Epac2-camps. Confocal imaging of di-8-ANNEPS-stained cells and immunoblotting were used to determine structural alterations. At 4-week post-MI, T-tubule regularity, density and junctophilin-2 (JPH2) expression were significantly decreased. The amplitude of local ß2AR-mediated cAMP in T-tubules was reduced and cAMP diffused throughout the cytosol instead of being locally confined. This was accompanied by partial caveolin-3 (Cav-3) dissociation from the membrane. At 8-week post-MI, the ß2AR-mediated cAMP response was observed at the T-tubules and the sarcolemma (crest). Finally, at 16-week post-MI, the whole cell ß2AR-mediated cAMP signal was depressed due to adenylate cyclase dysfunction, while overall Cav-3 levels were significantly increased and a substantial portion of Cav-3 dissociated into the cytosol. Overexpression of JPH2 in failing cells in vitro or AAV9.SERCA2a gene therapy in vivo did not improve ß2AR-mediated signal compartmentation or reduce cAMP diffusion. CONCLUSION: Although changes in T-tubule structure and ß2AR-mediated cAMP signalling are significant even at 4-week post-MI, progression to the HF phenotype is not linear. At 8-week post-MI the loss of ß2AR-mediated cAMP is temporarily reversed. Complete disorganization of ß2AR-mediated cAMP signalling due to changes in functional receptor localization and cellular structure occurs at 16-week post-MI.


Assuntos
AMP Cíclico/metabolismo , Insuficiência Cardíaca/metabolismo , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Sarcolema/metabolismo , Sistemas do Segundo Mensageiro , Remodelação Ventricular , Adenilil Ciclases/metabolismo , Animais , Técnicas Biossensoriais , Caveolina 3/metabolismo , Células Cultivadas , Difusão , Modelos Animais de Doenças , Progressão da Doença , Terapia Genética/métodos , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Eletroquímica de Varredura/métodos , Infarto do Miocárdio/complicações , Miócitos Cardíacos/patologia , Transporte Proteico , Ratos Sprague-Dawley , Sarcolema/patologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Fatores de Tempo , Transfecção
15.
Cardiovasc Res ; 110(2): 238-48, 2016 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-26956799

RESUMO

AIMS: Heart failure is often preceded by cardiac hypertrophy, which is characterized by increased cell size, altered protein abundance, and actin cytoskeletal reorganization. Profilin is a well-conserved, ubiquitously expressed, multifunctional actin-binding protein, and its role in cardiomyocytes is largely unknown. Given its involvement in vascular hypertrophy, we aimed to test the hypothesis that profilin-1 is a key mediator of cardiomyocyte-specific hypertrophic remodelling. METHODS AND RESULTS: Profilin-1 was elevated in multiple mouse models of hypertrophy, and a cardiomyocyte-specific increase of profilin in Drosophila resulted in significantly larger heart tube dimensions. Moreover, adenovirus-mediated overexpression of profilin-1 in neonatal rat ventricular myocytes (NRVMs) induced a hypertrophic response, measured by increased myocyte size and gene expression. Profilin-1 silencing suppressed the response in NRVMs stimulated with phenylephrine or endothelin-1. Mechanistically, we found that profilin-1 regulates hypertrophy, in part, through activation of the ERK1/2 signalling cascade. Confocal microscopy showed that profilin localized to the Z-line of Drosophila myofibrils under normal conditions and accumulated near the M-line when overexpressed. Elevated profilin levels resulted in elongated sarcomeres, myofibrillar disorganization, and sarcomeric disarray, which correlated with impaired muscle function. CONCLUSION: Our results identify novel roles for profilin as an important mediator of cardiomyocyte hypertrophy. We show that overexpression of profilin is sufficient to induce cardiomyocyte hypertrophy and sarcomeric remodelling, and silencing of profilin attenuates the hypertrophic response.


Assuntos
Cardiomegalia/genética , Cardiomegalia/metabolismo , Miócitos Cardíacos/metabolismo , Profilinas/genética , Profilinas/metabolismo , Animais , Drosophila melanogaster , Endotelina-1/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miofibrilas/metabolismo , Fenilefrina/farmacologia , Sarcômeros/metabolismo
16.
Stem Cells ; 34(1): 34-43, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26352327

RESUMO

Heart failure is still a major cause of hospitalization and mortality in developed countries. Many clinical trials have tested the use of multipotent stem cells as a cardiac regenerative medicine. The benefit for the patients of this therapeutic intervention has remained limited. Herein, we review the pluripotent stem cells as a cell source for cardiac regeneration. We more specifically address the various challenges of this cell therapy approach. We question the cell delivery systems, the immune tolerance of allogenic cells, the potential proarrhythmic effects, various drug mediated interventions to facilitate cell grafting and, finally, we describe the pathological conditions that may benefit from such an innovative approach. As members of a transatlantic consortium of excellence of basic science researchers and clinicians, we propose some guidelines to be applied to cell types and modes of delivery in order to translate pluripotent stem cell cardiac derivatives into safe and effective clinical trials.


Assuntos
Insuficiência Cardíaca/terapia , Miocárdio/citologia , Células-Tronco Pluripotentes/citologia , Transplante de Células-Tronco , Animais , Diferenciação Celular , Modelos Animais de Doenças , Insuficiência Cardíaca/fisiopatologia , Humanos
17.
PLoS One ; 10(5): e0126860, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25993131

RESUMO

Huntington disease (HD; OMIM 143100), a progressive neurodegenerative disorder, is caused by an expanded trinucleotide CAG (polyQ) motif in the HTT gene. Cardiovascular symptoms, often present in early stage HD patients, are, in general, ascribed to dysautonomia. However, cardio-specific expression of polyQ peptides caused pathological response in murine models, suggesting the presence of a nervous system-independent heart phenotype in HD patients. A positive correlation between the CAG repeat size and severity of symptoms observed in HD patients has also been observed in in vitro HD cellular models. Here, we test the suitability of human embryonic stem cell (hESC) lines carrying HD-specific mutation as in vitro models for understanding molecular mechanisms of cardiac pathology seen in HD patients. We have differentiated three HD-hESC lines into cardiomyocytes and investigated CAG stability up to 60 days after starting differentiation. To assess CAG stability in other tissues, the lines were also subjected to in vivo differentiation into teratomas for 10 weeks. Neither directed differentiation into cardiomyocytes in vitro nor in vivo differentiation into teratomas, rich in immature neuronal tissue, led to an increase in the number of CAG repeats. Although the CAG stability might be cell line-dependent, induced pluripotent stem cells generated from patients with larger numbers of CAG repeats could have an advantage as a research tool for understanding cardiac symptoms of HD patients.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias Humanas/metabolismo , Doença de Huntington/patologia , Mutação/genética , Miócitos Cardíacos/citologia , Expansão das Repetições de Trinucleotídeos/genética , Linhagem Celular , Humanos , Células-Tronco Pluripotentes/citologia
18.
Stem Cells Dev ; 24(7): 869-78, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25387407

RESUMO

Vascular derivatives of human embryonic stem cells (hESC) are being developed as sources of tissue-specific cells for organ regeneration. However, identity of developmental pathways that modulate the specification of endothelial cells is not known yet. We studied phosphatidylinositol 3-kinase (PI3K)-Forkhead box O transcription factor 1A (FOXO1A) pathways during differentiation of hESC toward endothelial lineage and on proliferation, maturation, and cell death of hESC-derived endothelial cells (hESC-EC). During differentiation of hESC, expression of FOXO1A transcription factor was linked to the expression of a cluster of angiogenesis- and vascular remodeling-related genes. PI3K inhibitor LY294002 activated FOXO1A and induced formation of CD31(+) hESC-EC. In contrast, differentiating hESC with silenced FOXO1A by small interfering RNA (siRNA) showed lower mRNA levels of CD31 and angiopoietin2. LY294002 decreased proliferative activity of purified hESC-EC, while FOXO1A siRNA increased their proliferation. LY294002 inhibits migration and tube formation of hESC-EC; in contrast, FOXO1A siRNA increased in vitro tube formation activity of hESC-EC. After in vivo conditioning of cells in athymic nude rats, cells retain their low FOXO1A expression levels. PI3K/FOXO1A pathway is important for function and survival of hESC-EC and in the regulation of endothelial cell fate. Understanding these properties of hESC-EC may help in future applications for treatment of injured organs.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Células-Tronco Embrionárias/citologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Neovascularização Fisiológica , Fosfatidilinositol 3-Quinases/genética , Ratos
20.
PLoS One ; 9(4): e91119, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24690886

RESUMO

Human embryonic stem cell-derived endothelial cells (hESC-EC), as well as other stem cell derived endothelial cells, have a range of applications in cardiovascular research and disease treatment. Endothelial cells sense Gram-negative bacteria via the pattern recognition receptors (PRR) Toll-like receptor (TLR)-4 and nucleotide-binding oligomerisation domain-containing protein (NOD)-1. These pathways are important in terms of sensing infection, but TLR4 is also associated with vascular inflammation and atherosclerosis. Here, we have compared TLR4 and NOD1 responses in hESC-EC with those of endothelial cells derived from other stem cells and with human umbilical vein endothelial cells (HUVEC). HUVEC, endothelial cells derived from blood progenitors (blood outgrowth endothelial cells; BOEC), and from induced pluripotent stem cells all displayed both a TLR4 and NOD1 response. However, hESC-EC had no TLR4 function, but did have functional NOD1 receptors. In vivo conditioning in nude rats did not confer TLR4 expression in hESC-EC. Despite having no TLR4 function, hESC-EC sensed Gram-negative bacteria, a response that was found to be mediated by NOD1 and the associated RIP2 signalling pathways. Thus, hESC-EC are TLR4 deficient but respond to bacteria via NOD1. This data suggests that hESC-EC may be protected from unwanted TLR4-mediated vascular inflammation, thus offering a potential therapeutic advantage.


Assuntos
Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Haemophilus influenzae/fisiologia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/microbiologia , Proteína Adaptadora de Sinalização NOD1/metabolismo , Animais , Células Endoteliais/citologia , Técnicas de Silenciamento de Genes , Infecções por Haemophilus/microbiologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/microbiologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos Nus , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Transplante de Células-Tronco , Receptor 4 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA