Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 318(2): C380-C391, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31913702

RESUMO

Children surviving cancer and chemotherapy are at risk for adverse health events including heart failure that may be delayed by years. Although the early effects of doxorubicin-induced cardiotoxicity may be attributed to a direct effect on the cardiomyocytes, the mechanisms underlying the delayed or late effects (8-20 yr) are unknown. The goal of this project was to develop a model of late-onset doxorubicin-induced cardiotoxicity to better delineate the underlying pathophysiology responsible. The underlying hypothesis was that doxorubicin-induced "late-onset cardiotoxicity" was the result of mitochondrial dysfunction leading to cell failure and death. Wistar rats, 3-4 wk of age, were randomly assigned to vehicle or doxorubicin injection groups (1-45 mg/kg). Cardiovascular function was unaltered at the lower dosages (1-15 kg/mg), but beginning at 6 mo after injection significant cardiac degradation was observed in the 45 mg/kg group. Doxorubicin significantly increased myocardial mitochondrial DNA (mtDNA) damage. In contrast, in isolated c-kit left ventricular (LV) cells, doxorubicin treatment did not increase mtDNA damage. Biomarkers of senescence within the LV were significantly increased, suggesting accelerated aging of the LV. Doxorubicin also significantly increased LV histamine content suggestive of mast cell activation. With the use of flow cytometry, a significant expansion of the c-kit and stage-specific embryonic antigen 1 cell populations within the LV were concomitant with significant decreases in the circulating peripheral blood population of these cells. These results are consistent with the concept that doxorubicin induced significant damage to the cardiomyocyte population and that although the heart attempted to compensate it eventually succumbed to an inability for self-repair.


Assuntos
Cardiotoxicidade/patologia , Senescência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Animais , Linhagem Celular , DNA Mitocondrial/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Doenças Mitocondriais/induzido quimicamente , Doenças Mitocondriais/patologia , Ratos , Ratos Wistar
3.
Am J Physiol Heart Circ Physiol ; 314(1): H68-H81, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28939651

RESUMO

Notch receptor signaling is active during cardiac development and silenced in myocytes after birth. Conversely, outward K+ Kv currents progressively appear in postnatal myocytes leading to shortening of the action potential (AP) and acquisition of the mature electrical phenotype. In the present study, we tested the possibility that Notch signaling modulates the electrical behavior of cardiomyocytes by interfering with Kv currents. For this purpose, the effects of Notch receptor activity on electrophysiological properties of myocytes were evaluated using transgenic mice with inducible expression of the Notch1 intracellular domain (NICD), the functional fragment of the activated Notch receptor, and in neonatal myocytes after inhibition of the Notch transduction pathway. By patch clamp, NICD-overexpressing cells presented prolonged AP duration and reduced upstroke amplitude, properties that were coupled with reduced rapidly activating Kv and fast Na+ currents, compared with cells obtained from wild-type mice. In cultured neonatal myocytes, inhibition of the proteolitic release of NICD with a γ-secretase antagonist increased transcript levels of the Kv channel-interacting proteins 2 (KChIP2) and enhanced the density of Kv currents. Collectively, these results indicate that Notch signaling represents an important regulator of the electrophysiological behavior of developing and adult myocytes by repressing, at least in part, repolarizing Kv currents. NEW & NOTEWORTHY We investigated the effects of Notch receptor signaling on the electrical properties of cardiomyocytes. Our results indicate that the Notch transduction pathway interferes with outward K+ Kv currents, critical determinants of the electrical repolarization of myocytes.


Assuntos
Miócitos Cardíacos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Potássio/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Feminino , Cinética , Proteínas Interatuantes com Canais de Kv/genética , Proteínas Interatuantes com Canais de Kv/metabolismo , Masculino , Potenciais da Membrana , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Receptor Notch1/genética , Sódio/metabolismo
4.
Stem Cells Int ; 2017: 3464953, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28791052

RESUMO

Previously, we reported that treatment with the G9a histone methyltransferase inhibitor BIX01294 causes bone marrow mesenchymal stem cells (MSCs) to exhibit a cardiocompetent phenotype, as indicated by the induction of the precardiac markers Mesp1 and brachyury. Here, we report that combining the histone deacetylase inhibitor trichostatin A (TSA) with BIX01294 synergistically enhances MSC cardiogenesis. Although TSA by itself had no effect on cardiac gene expression, coaddition of TSA to MSC cultures enhanced BIX01294-induced levels of Mesp1 and brachyury expression 5.6- and 7.2-fold. Moreover, MSCs exposed to the cardiogenic stimulus Wnt11 generated 2.6- to 5.6-fold higher levels of the cardiomyocyte markers GATA4, Nkx2.5, and myocardin when pretreated with TSA in addition to BIX01294. MSC cultures also showed a corresponding increase in the prevalence of sarcomeric protein-positive cells when treated with these small molecule inhibitors. These results correlated with data showing synergism between (1) TSA and BIX01294 in promoting acetylation of lysine 27 on histone H3 and (2) BIX01294 and Wnt11 in decreasing ß-catenin accumulation in MSCs. The implications of these findings are discussed in light of observations in the early embryo on the importance of ß-catenin signaling and histone modifications for cardiomyocyte differentiation and heart development.

5.
Stem Cells Int ; 2015: 270428, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26089912

RESUMO

The G9a histone methyltransferase inhibitor BIX01294 was examined for its ability to expand the cardiac capacity of bone marrow cells. Inhibition of G9a histone methyltransferase by gene specific knockdown or BIX01294 treatment was sufficient to induce expression of precardiac markers Mesp1 and brachyury in bone marrow cells. BIX01294 treatment also allowed bone marrow mesenchymal stem cells (MSCs) to express the cardiac transcription factors Nkx2.5, GATA4, and myocardin when subsequently exposed to the cardiogenic stimulating factor Wnt11. Incubation of BIX01294-treated MSCs with cardiac conditioned media provoked formation of phase bright cells that exhibited a morphology and molecular profile resembling similar cells that normally form from cultured atrial tissue. Subsequent aggregation and differentiation of BIX01294-induced, MSC-derived phase bright cells provoked their cardiomyogenesis. This latter outcome was indicated by their widespread expression of the primary sarcomeric proteins muscle α-actinin and titin. MSC-derived cultures that were not initially treated with BIX01294 exhibited neither a commensurate burst of phase bright cells nor stimulation of sarcomeric protein expression. Collectively, these data indicate that BIX01294 has utility as a pharmacological agent that could enhance the ability of an abundant and accessible stem cell population to regenerate new myocytes for cardiac repair.

6.
Cell Reprogram ; 16(5): 324-30, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25090621

RESUMO

The DNA methylation inhibitor 5-azacytidine is widely used to stimulate the cardiac differentiation of stem cells. However, 5-azacytidine has long been employed as a tool for stimulating skeletal myogenesis. Yet, it is unclear whether the ability of 5-azacytidine to promote both cardiac and skeletal myogenesis is dependent strictly on the native potential of the starting cell population or if this drug is a transdifferentiation agent. To address this issue, we examined the effect of 5-azacytidine on cultures of adult mouse atrial tissue, which contains cardiac but not skeletal muscle progenitors. Exposure to 5-azacytidine caused atrial cells to elongate and increased the presence of fat globules within the cultures. 5-Azacytidine also induced expression of the skeletal myogenic transcription factors MyoD and myogenin. 5-Azacytidine pretreatments allowed atrial cells to undergo adipogenesis or skeletal myogenesis when subsequently cultured with either insulin and dexamethasone or low-serum media, respectively. The presence of skeletal myocytes in atrial cultures was indicated by dual staining for myogenin and sarcomeric α-actin. These data demonstrate that 5-azacytidine converts cardiac cells to noncardiac cell types and suggests that this drug has a compromised efficacy as a cardiac differentiation factor.


Assuntos
Azacitidina/farmacologia , Transdiferenciação Celular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Miocárdio/citologia , Animais , Linhagem Celular , Camundongos , Camundongos Endogâmicos BALB C , Músculo Esquelético/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Biotechnol Lett ; 35(10): 1707-14, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23690049

RESUMO

Chronic exposure to solar radiation is the primary cause of photoaging and benign and malignant skin tumors. A conditioned serum-free medium (SFM) was prepared from umbilical cord mesenchymal stem cells (UC-MSCs) and its anti-photoaging effect, following chronic UV irradiation in vitro and in vivo, was evaluated. UC-MSC SFM had a stimulatory effect on human dermal fibroblast proliferation and reduced UVA-induced cell death. In addition, UC-MSC SFM blocked UVA inhibition of superoxide dismutase activity. Topical application of UC-MSC SFM to mouse skin prior to UV irradiation blocked the inhibition of superoxide dismutase and glutathione peroxidase activities, and prevented the upregulation of malonaldehyde. UC-MSC SFM thus protects against photoaging induced by UVA and UVB radiation and is a promising candidate for skin anti-photoaging treatments.


Assuntos
Sobrevivência Celular/efeitos da radiação , Meios de Cultivo Condicionados/química , Fibroblastos/efeitos da radiação , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Raios Ultravioleta , Cordão Umbilical/citologia , Animais , Proliferação de Células , Células Cultivadas , Meios de Cultura Livres de Soro/química , Fibroblastos/fisiologia , Glutationa Peroxidase/metabolismo , Humanos , Malondialdeído/metabolismo , Camundongos , Pele/enzimologia , Pele/metabolismo , Pele/efeitos da radiação , Superóxido Dismutase/metabolismo
8.
PLoS One ; 8(2): e56554, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23418583

RESUMO

Understanding how stem cells interact with cardiomyocytes is crucial for cell-based therapies to restore the cardiomyocyte loss that occurs during myocardial infarction and other cardiac diseases. It has been thought that functional myocardial repair and regeneration could be regulated by stem cell-cardiomyocyte contact. However, because various contact modes (junction formation, cell fusion, partial cell fusion, and tunneling nanotube formation) occur randomly in a conventional coculture system, the particular regulation corresponding to a specific contact mode could not be analyzed. In this study, we used laser-patterned biochips to define cell-cell contact modes for systematic study of contact-mediated cellular interactions at the single-cell level. The results showed that the biochip design allows defined stem cell-cardiomyocyte contact-mode formation, which can be used to determine specific cellular interactions, including electrical coupling, mechanical coupling, and mitochondria transfer. The biochips will help us gain knowledge of contact-mediated interactions between stem cells and cardiomyocytes, which are fundamental for formulating a strategy to achieve stem cell-based cardiac tissue regeneration.


Assuntos
Comunicação Celular/fisiologia , Rastreamento de Células/métodos , Células-Tronco Mesenquimais/fisiologia , Miócitos Cardíacos/fisiologia , Animais , Animais Recém-Nascidos , Fusão Celular , Membrana Celular/fisiologia , Rastreamento de Células/instrumentação , Células Cultivadas , Técnicas de Cocultura , Corantes Fluorescentes/química , Imuno-Histoquímica , Indóis/química , Junções Intercelulares/fisiologia , Lasers , Células-Tronco Mesenquimais/citologia , Microscopia Confocal , Microscopia de Fluorescência , Mitocôndrias/fisiologia , Miócitos Cardíacos/citologia , Ratos , Reprodutibilidade dos Testes , Análise de Célula Única/instrumentação , Análise de Célula Única/métodos
9.
Artigo em Inglês | MEDLINE | ID: mdl-24527266

RESUMO

Mesenchymal stem cells (MSCs) have been cited as contributors to heart repair through cardiogenic differentiation and multiple cellular interactions, including the paracrine effect, cell fusion, and mechanical and electrical couplings. Due to heart-muscle complexity, progress in the development of knowledge concerning the role of MSCs in cardiac repair is heavily based on MSC-cardiomyocyte coculture. In conventional coculture systems, however, the in vivo cardiac muscle structure, in which rod-shaped cells are connected end-to-end, is not sustained; instead, irregularly shaped cells spread randomly, resulting in randomly distributed cell junctions. Consequently, contact-mediated cell-cell interactions (e.g., the electrical triggering signal and the mechanical contraction wave that propagate through MSC-cardiomyocyte junctions) occur randomly. Thus, the data generated on the beneficial effects of MSCs may be irrelevant to in vivo biological processes. In this study, we explored whether cardiomyocyte alignment, the most important phenotype, is relevant to stem cell cardiogenic differentiation. Here, we report (i) the construction of a laser-patterned, biochip-based, stem cell-cardiomyocyte coculture model with controlled cell alignment; and (ii) single-cell-level data on stem cell cardiogenic differentiation under in vivo-like cardiomyocyte alignment conditions.

10.
Stem Cells Dev ; 22(4): 654-67, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22994322

RESUMO

Bone marrow (BM) has long been considered a potential stem cell source for cardiac repair due to its abundance and accessibility. Although previous investigations have generated cardiomyocytes from BM, yields have been low, and far less than produced from ES or induced pluripotent stem cells (iPSCs). Since differentiation of pluripotent cells is difficult to control, we investigated whether BM cardiac competency could be enhanced without making cells pluripotent. From screens of various molecules that have been shown to assist iPSC production or maintain the ES cell phenotype, we identified the G9a histone methyltransferase inhibitor BIX01294 as a potential reprogramming agent for converting BM cells to a cardiac-competent phenotype. BM cells exposed to BIX01294 displayed significantly elevated expression of brachyury, Mesp1, and islet1, which are genes associated with embryonic cardiac progenitors. In contrast, BIX01294 treatment minimally affected ectodermal, endodermal, and pluripotency gene expression by BM cells. Expression of cardiac-associated genes Nkx2.5, GATA4, Hand1, Hand2, Tbx5, myocardin, and titin was enhanced 114, 76, 276, 46, 635, 123, and 5-fold in response to the cardiogenic stimulator Wnt11 when BM cells were pretreated with BIX01294. Immunofluorescent analysis demonstrated that BIX01294 exposure allowed for the subsequent display of various muscle proteins within the cells. The effect of BIX01294 on the BM cell phenotype and differentiation potential corresponded to an overall decrease in methylation of histone H3 at lysine9, which is the primary target of G9a histone methyltransferase. In summary, these data suggest that BIX01294 inhibition of chromatin methylation reprograms BM cells to a cardiac-competent progenitor phenotype.


Assuntos
Azepinas/farmacologia , Células da Medula Óssea , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Miocárdio , Miócitos Cardíacos , Quinazolinas/farmacologia , Animais , Antígenos de Diferenciação/biossíntese , Células da Medula Óssea/citologia , Células da Medula Óssea/enzimologia , Regulação da Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/metabolismo , Camundongos , Proteínas Musculares/metabolismo , Miocárdio/citologia , Miocárdio/enzimologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/enzimologia
11.
Cell Mol Bioeng ; 5(3): 327-336, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23139730

RESUMO

Normal cardiomyocytes are highly dependent on the functional expression of ion channels to form action potentials and electrical coupling with other cells. To fully determine the scientific and therapeutic potential of stem cells for cardiovascular-disease treatment, it is necessary to assess comprehensively the regulation of stem-cell electrical properties during stem cell-cardiomyocyte interaction. It has been reported in the literature that contact with native cardiomyocytes induced and regulated stem-cell cardiogenic differentiation. However, in conventional cell-culture models, the importance of cell-cell contact for stem-cell functional coupling with cardiomyocytes has not been elucidated due to insufficient control of the cell-contact mode of individual cells. Using microfabrication and laser-guided cell micropatterning techniques, we created two biochips with contact-promotive and -preventive microenvironments to systematically study the effect of contact on cardiogenic regulation of stem-cell electrical properties. In contact-promotive biochips, connexin 43 expression was upregulated and relocated to the junction area between one stem cell and one cardiomyocyte. Only stem cells in contact with cardiomyocytes were induced by adjacent cardiomyocytes to acquire electrophysiological properties for action-potential formation similar to that of a cardiomyocyte.

12.
Dev Growth Differ ; 54(2): 153-66, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22150286

RESUMO

Lithium is a commonly used drug for the treatment of bipolar disorder. At high doses, lithium becomes teratogenic, which is a property that has allowed this agent to serve as a useful tool for dissecting molecular pathways that regulate embryogenesis. This study was designed to examine the impact of lithium on heart formation in the developing frog for insights into the molecular regulation of cardiac specification. Embryos were exposed to lithium at the beginning of gastrulation, which produced severe malformations of the anterior end of the embryo. Although previous reports characterized this deformity as a posteriorized phenotype, histological analysis revealed that the defects were more comprehensive, with disfigurement and disorganization of all interior tissues along the anterior-posterior axis. Emerging tissues were poorly segregated and cavity formation was decreased within the embryo. Lithium exposure also completely ablated formation of the heart and prevented myocardial cell differentiation. Despite the complete absence of cardiac tissue in lithium treated embryos, exposure to lithium did not prevent myocardial differentiation of precardiac dorsal marginal zone explants. Moreover, precardiac tissue freed from the embryo subsequent to lithium treatment at gastrulation gave rise to cardiac tissue, as demonstrated by upregulation of cardiac gene expression, display of sarcomeric proteins, and formation of a contractile phenotype. Together these data indicate that lithium's effect on the developing heart was not due to direct regulation of cardiac differentiation, but an indirect consequence of disrupted tissue organization within the embryo.


Assuntos
Embrião não Mamífero/efeitos dos fármacos , Coração/embriologia , Lítio/farmacologia , Animais , Embrião não Mamífero/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Xenopus laevis
13.
Am J Physiol Heart Circ Physiol ; 301(5): H1952-64, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21908788

RESUMO

This study examined transgenic mice whose expression of a ß-galactosidase (lacZ) reporter is driven by a GATA6 gene enhancer. Previous investigations established that transcription of the transgene was associated with precardiac mesoderm and primary heart tube myocardium, which decreased progressively, so that its expression was no longer observed within ventricular myocardium by midgestation. Expression of this reporter in the adult was investigated for insights into myocyte homeostasis and cardiovascular biology. Morphometric analysis determined that <1% of myocytes, often found in small clusters, express this GATA6-associated reporter in the adult heart. LacZ expression was also found in the ascending aorta. Myocardial expression of the transgene was not associated with a proliferative phenotype or new myocyte formation, as lacZ-positive myocytes neither labeled with cell division markers nor following 5-bromodeoxyuridine pulse-chase experimentation. Despite exhibiting normal adherens junctions, these myocytes appeared to exhibit decreased connexin 43 gap junctions. Treatment with the gap junctional blocker heptanol both in vivo and in culture elevated myocardial ß-galactosidase activity, suggesting that deficient gap junctional communication underlies expression of the transgenic reporter. LacZ expression within the myocardium was also enhanced in response to cryoinjury and isoproterenol-induced hypertrophy. These results reveal a previously uncharacterized phenotypic heterogeneity in the myocardium and suggest that decreased gap junctional coupling leads to induction of a signaling pathway that utilizes a unique GATA6 enhancer. Upregulation of lacZ reporter gene expression following cardiac injury indicates this transgenic mouse may serve as a model for examining the transition of the heart from healthy to pathological states.


Assuntos
Comunicação Celular/genética , Fator de Transcrição GATA6/genética , Junções Comunicantes/metabolismo , Genes Reporter , Óperon Lac , Miócitos Cardíacos/metabolismo , Regiões Promotoras Genéticas , Junções Aderentes/metabolismo , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Comunicação Celular/efeitos dos fármacos , Células Cultivadas , Conexina 43/metabolismo , Modelos Animais de Doenças , Junções Comunicantes/efeitos dos fármacos , Genótipo , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Heptanol/farmacologia , Isoproterenol , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fenótipo , Regulação para Cima , beta-Galactosidase/biossíntese , beta-Galactosidase/genética
14.
Stem Cells Dev ; 20(11): 1973-83, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21351874

RESUMO

WNT signaling has been shown to influence the development of the heart. Although recent data suggested that canonical WNTs promote the emergence and expansion of cardiac progenitors in the pregastrula embryo, it has long been accepted that once gastrulation begins, canonical WNT signaling needs to be suppressed for cardiac development to proceed. Yet, this latter supposition appears to be odds with the expression of multiple canonical WNTs in the developing heart. The present study examining the effect of ectopic canonical WNT signaling on cardiogenesis in the developing frog was designed to test the hypothesis that heart formation is dependent on the inhibition of canonical WNT activity at the onset of gastrulation. Here we report that cardiac differentiation of explanted precardiac tissue from the dorsal marginal zone was not suppressed by exposure to WNT1 protein, although expression of Tbx5, Tbx20, and Nkx2.5 was selectively reduced. Pharmacological activation of WNT signaling in intact embryos using the GSK3 inhibitor SB415286 did not prevent the formation of an anatomically normal and functionally sound heart, with the only defect observed being lower levels of the cardiac transcription factor Nkx2.5. In both the explant and whole embryo studies, expression of muscle genes and proteins was unaffected by ectopic canonical WNT signaling. In contrast, canonical Wnt signaling upregulated expression of the cardiac stem cell marker c-kit and pluripotency genes Oct25 and Oct60. However, this regulatory stimulation of stem cells did not come at the expense of blocking cardiac progenitors from differentiating.


Assuntos
Diferenciação Celular , Coração/crescimento & desenvolvimento , Larva/crescimento & desenvolvimento , Miocárdio/citologia , Transdução de Sinais , Células-Tronco/fisiologia , Via de Sinalização Wnt , Xenopus laevis/crescimento & desenvolvimento , Aminofenóis/farmacologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Blástula/citologia , Blástula/metabolismo , Feminino , Gastrulação , Expressão Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Larva/genética , Larva/metabolismo , Maleimidas/farmacologia , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Sarcômeros/metabolismo , Células-Tronco/metabolismo , Técnicas de Cultura de Tecidos , Proteína Wnt1/farmacologia , Proteína Wnt1/fisiologia , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo
15.
ScientificWorldJournal ; 7: 161-76, 2007 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-17334608

RESUMO

Wnts are a family of secreted signaling proteins that are encoded by 19 distinct genes in the vertebrate genome. These molecules initiate several signal transduction pathways: the canonical Wnt, Wnt/Ca2+, and Wnt/planar cell polarity pathways. Wnt proteins have major impact on embryonic development, tumor progression, and stem cell differentiation. Wnt signal transduction also influences the formation of the heart, yet many issues concerning the involvement of Wnt regulation in initiating cardiac development remain unresolved. In this review, we will examine the published record to discern (a) what has been shown by experimental studies on the participation of Wnt signaling in cardiogenesis, and (b) what are the important questions that need to be addressed to understand the importance and function of Wnt signal transduction in facilitating the development of the heart.


Assuntos
Coração/embriologia , Coração/fisiologia , Mesoderma/fisiologia , Organogênese/fisiologia , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , Animais , Humanos
16.
Tissue Eng ; 12(4): 853-65, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16674298

RESUMO

A major aim of regenerative medicine is the construction of bioengineered organs and tissue for transplantation into human patients; yet living tissue is dynamic, and thus arranging cellular and extracellular constituents into an architecture resembling normal adult organs may not be sufficient to maintain tissue stability. In this study, we used cultures of embryonic chick heart tissue as a model to explore how newly formed cardiac tissue constructs can sustain their morphological structure and functional capabilities over extended periods. During the initial days of incubation, embryonic cardiac explants will thrive as beating three-dimensional tissue aggregates. However, within the first week of culture, cardiac aggregates lose their contractile function and flatten. After 2 weeks of incubation, the cardiac cells will have spread out into a homogeneous monolayer and dedifferentiated to a noncardiac phenotype. In contrast, when the embryonic heart tissue was co-cultured with a noncardiac cell layer obtained from adult bone marrow, the cardiac aggregates maintained their contractile function, three-dimensional tissue morphology, and myocyte phenotype for a full month of incubation. The capacity of this noncardiac cell layer to sustain the phenotype and morphology of the cardiac explants was partially replicated by treatment of the heart tissue with conditioned media from bone marrow cells. These findings are discussed in regard to the importance of adjacent cell layers for facilitating organogenesis in the developing embryo and having potential utility in producing stable bioengineered tissue constructs.


Assuntos
Células da Medula Óssea/fisiologia , Longevidade/fisiologia , Miocárdio/citologia , Miocárdio/metabolismo , Engenharia Tecidual/métodos , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Rim/citologia , Camundongos , Camundongos Endogâmicos ICR , Técnicas de Cultura de Órgãos , Fatores de Tempo
17.
Dev Biol ; 293(2): 305-15, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16563368

RESUMO

Soon after fertilization, vertebrate embryos grow very rapidly. Thus, early in gestation, a sizeable yet underdeveloped organism requires circulating blood. This need dictates the early appearance of a contractile heart, which is the first functional organ in both the avian and mammalian embryo. The heart arises from paired mesodermal regions within the anterior half of the embryo. As development proceeds, these bilateral precardiac fields merge at the midline to give rise to the primary heart tube. How specific areas of nondifferentiated mesoderm organize into myocardial tissue has been a question that has long intrigued developmental biologists. In recent years, the regulation of Wnt signal transduction has been implicated as an important event that initiates cardiac development. While initial reports in Drosophila and the bird had implicated Wnt proteins as promoters of cardiac tissue formation, subsequent findings that the WNT inhibitors Dkk1 and crescent possess cardiac-inducing activities led to the contrary hypothesis that WNTs actively inhibit cardiogenesis. This seeming contradiction has been resolved, in part, by more recent information indicating that Wnts stimulate multiple signal transduction pathways. In this review, we will examine what is presently known about the importance of regulated Wnt activity for the formation of the heart and the development of the myocardium and discuss this information in context of the emerging complexity of Wnt signal transduction.


Assuntos
Coração Fetal/embriologia , Proteínas Wnt/fisiologia , Animais , Coração Fetal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Modelos Biológicos , Transdução de Sinais , Vertebrados/embriologia , Vertebrados/genética , Vertebrados/fisiologia , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/genética
18.
Stem Cells ; 24(5): 1236-45, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16410395

RESUMO

Since rates of cardiomyocyte generation in the embryo are much higher than within the adult, we explored whether the embryonic heart would serve as useful experimental system for examining the myocardial potential of adult stem cells. Previously, we reported that the long-term culturing of adult mouse bone marrow produced a cell population that was both highly enriched for macrophages and cardiac competent. In this study, the myocardial potential of this cell population was analyzed in greater detail using the embryonic chick heart as recipient tissue. Experiments involving the co-incubation of labeled bone marrow cells with embryonic heart tissue showed that bone marrow (BM) cells incorporated into the myocardium and immunostained for myocyte proteins. Reverse transcription-polymerase chain reaction analysis demonstrated that the heart tissue induced bone marrow cells to express the differentiated cardiomyocyte marker alpha-cardiac myosin heavy chain. The cardiomyocyte conversion of the bone marrow cells was verified by harvesting donor cells from mice that were genetically labeled with a myocardial-specific beta-galactosidase reporter. Embryonic hearts exposed to the transgenic bone marrow in culture exhibited significant numbers of beta-galactosidase-positive cells, indicating the presence of bone marrow-derived cells that had converted to a myocardial phenotype. Furthermore, when transgenic mouse BM cells were injected into living chick embryos, donor cells incorporated into the developing heart and exhibited a myocardial phenotype. Immunofluorescence analysis demonstrated that donor BM cells exhibiting myocyte markers contained only nuclei from mouse cells, indicating that differentiation and not cell fusion was the predominant mechanism for the acquisition of a myocyte phenotype. These data confirm that adult mouse bone marrow contain cells with the ability to form cardiomyocytes. In addition, the predominance of the macrophage phenotype within the donor bone marrow cell population suggests that transdifferentiation of immune response cells may play a role in cellular regeneration in the adult.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/fisiologia , Proliferação de Células , Coração/embriologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Animais , Transplante de Medula Óssea , Embrião de Galinha , Meios de Cultivo Condicionados , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Macrófagos/citologia , Camundongos , Camundongos Transgênicos , Transplante Heterólogo
19.
Anat Rec A Discov Mol Cell Evol Biol ; 276(1): 2-12, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14699629

RESUMO

A major goal in cardiovascular biology is to repair diseased or damaged hearts with newly generated myocardial tissue. Stem cells offer a potential source of replacement myocytes for restoring cardiac function. Yet little is known about the nature of the cells that are able to generate myocardium and the conditions they require to form heart tissue. A source of information that may be pertinent to addressing these issues is the study of how the myocardium arises from progenitor cells in the early vertebrate embryo. Accordingly, this review will examine the initial events of cardiac developmental biology for insights into the identity and characteristics of the stem cells that can be used to generate myocardial tissue for therapeutic purposes.


Assuntos
Coração/embriologia , Células-Tronco Multipotentes/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Animais , Embrião de Galinha , Camundongos , Modelos Animais , Células-Tronco Multipotentes/fisiologia , Miócitos Cardíacos/fisiologia
20.
Anat Rec A Discov Mol Cell Evol Biol ; 276(1): 103-12, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14699637

RESUMO

Adult cardiac muscle is unable to repair itself following severe disease or injury. Because of this fundamental property of the myocardium, it was long believed that the adult myocardium is a postmitotic tissue. Yet, recent studies have indicated that new cardiac myocytes are generated throughout the life span of an adult and that extracardiac cells can contribute to the renewal of individual cells within the myocardium. In addition, investigations of the phenotypic capacity of adult stem cells have suggested that their potential is not solely restricted to the differentiated cell phenotypes of the source tissue. These observations have great implications for cardiac biology, as stem cells obtained from the bone marrow and other readily accessible adult tissues may serve as a source of replacement cardiac myocytes. In this review, we describe the evidence for these new findings and discuss their implications in context of the continuing controversy over stem cell plasticity.


Assuntos
Células-Tronco Mesenquimais/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Adulto , Animais , Humanos , Células-Tronco Mesenquimais/fisiologia , Miócitos Cardíacos/fisiologia , Células-Tronco Pluripotentes/fisiologia , Regeneração/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...