Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Respir Res ; 24(1): 165, 2023 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-37344798

RESUMO

BACKGROUND: Pulmonary arterial hypertension (PAH) is a rare but fatal cardiopulmonary disease mainly characterized by pulmonary vascular remodeling. Aberrant expression of circRNAs has been reported to play a crucial role in pulmonary vascular remodeling. The existing literature predominantly centers on studies that examined the sponge mechanism of circRNAs. However, the mechanism of circRNAs in regulating PAH-related protein remains largely unknown. This study aimed to investigate the effect of circItgb5 on pulmonary vascular remodeling and the underlying functional mechanism. MATERIALS AND METHODS: High-throughput circRNAs sequencing was used to detect circItgb5 expression in control and PDGF-BB-treated pulmonary arterial smooth muscle cells (PASMCs). Localization of circItgb5 in PASMCs was determined via the fluorescence in situ hybridization assay. Sanger sequencing was applied to analyze the circularization of Itgb5. The identification of proteins interacting with circItgb5 was achieved through a RNA pull-down assay. To assess the impact of circItgb5 on PASMCs proliferation, an EdU assay was employed. Additionally, the cell cycle of PASMCs was examined using a flow cytometry assay. Western blotting was used to detect biomarkers associated with the phenotypic switch of PASMCs. Furthermore, a monocrotaline (MCT)-induced PAH rat model was established to explore the effect of silencing circItgb5 on pulmonary vascular remodeling. RESULTS: CircItgb5 was significantly upregulated in PDGF-BB-treated PASMCs and was predominately localized in the cytoplasm of PASMCs. In vivo experiments revealed that the knockdown of circItgb5 attenuated MCT-induced pulmonary vascular remodeling and right ventricular hypertrophy. In vitro experiments revealed that circItgb5 promoted the transition of PASMCs to synthetic phenotype. Mechanistically, circItgb5 sponged miR-96-5p to increase mTOR level and interacted with Uba1 protein to activate the Ube2n/Mdm2/ACE2 pathway. CONCLUSIONS: CircItgb5 promoted the transition of PASMCs to synthetic phenotype by interacting with miR-96-5p and Uba1 protein. Knockdown of circItgb5 mitigated pulmonary arterial pressure, pulmonary vascular remodeling and right ventricular hypertrophy. Overall, circItgb5 has the potential for application as a therapeutic target for PAH.


Assuntos
Hipertensão Pulmonar , Cadeias beta de Integrinas , RNA Circular , Animais , Masculino , Ratos , Células Cultivadas , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , MicroRNAs/metabolismo , Monocrotalina , Mioblastos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-sis , Ratos Sprague-Dawley , RNA Circular/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima , Remodelação Vascular , Cadeias beta de Integrinas/genética
2.
Cell Death Dis ; 10(3): 210, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30824685

RESUMO

Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease affecting both the upper and lower motor neurons (MNs), with no effective treatment currently available. Early pathological events in ALS include perturbations in axonal transport (AT), formation of toxic protein aggregates and Neuromuscular Junction (NMJ) disruption, which all lead to axonal degeneration and motor neuron death. Pridopidine is a small molecule that has been clinically developed for Huntington disease. Here we tested the efficacy of pridopidine for ALS using in vitro and in vivo models. Pridopidine beneficially modulates AT deficits and diminishes NMJ disruption, as well as motor neuron death in SOD1G93A MNs and in neuromuscular co-cultures. Furthermore, we demonstrate that pridopidine activates the ERK pathway and mediates its beneficial effects through the sigma-1 receptor (S1R). Strikingly, in vivo evaluation of pridopidine in SOD1G93A mice reveals a profound reduction in mutant SOD1 aggregation in the spinal cord, and attenuation of NMJ disruption, as well as subsequent muscle wasting. Taken together, we demonstrate for the first time that pridopidine improves several cellular and histological hallmark pathologies of ALS through the S1R.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Neurônios Motores/efeitos dos fármacos , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Receptores sigma/metabolismo , Medula Espinal/efeitos dos fármacos , Superóxido Dismutase-1/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Transporte Axonal/efeitos dos fármacos , Transporte Axonal/genética , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Feminino , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Neurônios Motores/metabolismo , Células Musculares/efeitos dos fármacos , Células Musculares/metabolismo , Células Musculares/patologia , Mioblastos de Músculo Liso , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/genética , Junção Neuromuscular/fisiologia , Receptores sigma/genética , Medula Espinal/metabolismo , Medula Espinal/patologia , Superóxido Dismutase-1/genética , Receptor Sigma-1
3.
Am J Physiol Lung Cell Mol Physiol ; 315(2): L328-L333, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29722559

RESUMO

Asthma is characterized by a chronic inflammation and remodeling of the airways. Although inflammation can be controlled, therapeutic options to revert remodeling do not exist. Thus, there is a large and unmet need to understand the underlying molecular mechanisms to develop novel therapies. We previously identified a pivotal role for miR-142-3p in regulating airway smooth muscle (ASM) precursor cell proliferation during lung development by fine-tuning the Wingless/Integrase I (WNT) signaling. Thus, we here aimed to investigate the relevance of this interaction in asthma. We performed quantitative RT-PCR and immune staining in a murine model for ovalbumin-induced allergic airway inflammation and in bronchial biopsies from patients with asthma and isolated primary fibroblasts thereof. miR-142-3p was increased in hyperproliferative regions of lung in murine and human asthma, whereas this microRNA (miRNA) was excluded from regions with differentiated ASM cells. Increases in miR-142-3p were associated with a decrease of its known target Adenomatous polyposis coli. Furthermore, we observed a differential expression of miR-142-3p in bronchial biopsies from patients with early or late onset severe asthma, which coincided with a differential WNT signature. Our data suggest that miR-142-3p is involved in regulating the balance between proliferation and differentiation of ASM cells in asthma, possibly via controlling WNT signaling. Thus, this miRNA might be an interesting target to prevent ASM hyperproliferation in asthma.


Assuntos
Remodelação das Vias Aéreas , Asma/metabolismo , MicroRNAs/biossíntese , Miócitos de Músculo Liso/metabolismo , Via de Sinalização Wnt , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Asma/patologia , Asma/fisiopatologia , Proliferação de Células , Feminino , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Mioblastos de Músculo Liso/metabolismo , Mioblastos de Músculo Liso/patologia , Miócitos de Músculo Liso/patologia
4.
Cell Rep ; 23(4): 1152-1165, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29694892

RESUMO

Pulmonary hypertension is a devastating disease characterized by excessive vascular muscularization. We previously demonstrated primed platelet-derived growth factor receptor ß+ (PDGFR-ß+)/smooth muscle cell (SMC) marker+ progenitors at the muscular-unmuscular arteriole border in the normal lung, and in hypoxia-induced pulmonary hypertension, a single primed cell migrates distally and expands clonally, giving rise to most of the pathological smooth muscle coating of small arterioles. Little is known regarding the molecular mechanisms underlying this process. Herein, we show that primed cell expression of Kruppel-like factor 4 and hypoxia-inducible factor 1-α (HIF1-α) are required, respectively, for distal migration and smooth muscle expansion in a sequential manner. In addition, the HIF1-α/PDGF-B axis in endothelial cells non-cell autonomously regulates primed cell induction, proliferation, and differentiation. Finally, myeloid cells transdifferentiate into or fuse with distal arteriole SMCs during hypoxia, and Pdgfb deletion in myeloid cells attenuates pathological muscularization. Thus, primed cell autonomous and non-cell autonomous pathways are attractive therapeutic targets for pulmonary hypertension.


Assuntos
Transdiferenciação Celular , Hipertensão Pulmonar/metabolismo , Músculo Liso Vascular/metabolismo , Mioblastos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Feminino , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Linfocinas/genética , Linfocinas/metabolismo , Masculino , Camundongos , Músculo Liso Vascular/patologia , Mioblastos de Músculo Liso/patologia , Miócitos de Músculo Liso/patologia , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo
5.
Mol Metab ; 6(11): 1429-1442, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29107290

RESUMO

OBJECTIVE: Strategies improving skeletal muscle mitochondrial capacity are commonly paralleled by improvements in (metabolic) health. We and others previously identified microRNAs regulating mitochondrial oxidative capacity, but data in skeletal muscle are limited. Therefore, the present study aimed to identify novel microRNAs regulating skeletal muscle mitochondrial metabolism. METHODS AND RESULTS: We conducted an unbiased, hypothesis-free microRNA silencing screen in C2C12 myoblasts, using >700 specific microRNA inhibitors, and investigated a broad panel of mitochondrial markers. After subsequent validation in differentiated C2C12 myotubes, and exclusion of microRNAs without a human homologue or with an adverse effect on mitochondrial metabolism, 19 candidate microRNAs remained. Human clinical relevance of these microRNAs was investigated by measuring their expression in human skeletal muscle of subject groups displaying large variation in skeletal muscle mitochondrial capacity. CONCLUSION: The results show that that microRNA-320a, microRNA-196b-3p, microRNA-150-5p, and microRNA-34c-3p are tightly related to in vivo skeletal muscle mitochondrial function in humans and identify these microRNAs as targets for improving mitochondrial metabolism.


Assuntos
MicroRNAs/genética , Mitocôndrias Musculares/metabolismo , Mioblastos de Músculo Liso/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Humanos , Masculino , Camundongos , MicroRNAs/metabolismo , Mitocôndrias Musculares/genética , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo
6.
Stem Cell Res Ther ; 8(1): 156, 2017 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-28676082

RESUMO

BACKGROUND: Smooth muscle progenitor cells (pSMCs) differentiated from human pluripotent stem cells (hPSCs) hold great promise for treating diseases or degenerative conditions involving smooth muscle pathologies. However, the therapeutic potential of pSMCs derived from men and women may be very different. Cell sex can exert a profound impact on the differentiation process of stem cells into somatic cells. In spite of advances in translation of stem cell technologies, the role of cell sex and the effect of sex hormones on the differentiation towards mesenchymal lineage pSMCs remain largely unexplored. METHODS: Using a standard differentiation protocol, two human embryonic stem cell lines (one male line and one female line) and three induced pluripotent stem cell lines (one male line and two female lines) were differentiated into pSMCs. We examined differences in the differentiation of male and female hPSCs into pSMCs, and investigated the effect of 17ß-estradiol (E2) on the extracellular matrix (ECM) metabolisms and cell proliferation rates of the pSMCs. Statistical analyses were performed by using Student's t test or two-way ANOVA, p < 0.05. RESULTS: Male and female hPSCs had similar differentiation efficiencies and generated morphologically comparable pSMCs under a standard differentiation protocol, but the derived pSMCs showed sex differences in expression of ECM proteins, such as MMP-2 and TIMP-1, and cell proliferation rates. E2 treatment induced the expression of myogenic gene markers and suppressed ECM degradation activities through reduction of MMP activity and increased expression of TIMP-1 in female pSMCs, but not in male pSMCs. CONCLUSIONS: hPSC-derived pSMCs from different sexes show differential expression of ECM proteins and proliferation rates. Estrogen appears to promote maturation and ECM protein expression in female pSMCs, but not in male pSMCs. These data suggest that intrinsic cell-sex differences may influence progenitor cell biology.


Assuntos
Diferenciação Celular , Proliferação de Células , Proteínas da Matriz Extracelular/biossíntese , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Mioblastos de Músculo Liso/metabolismo , Caracteres Sexuais , Feminino , Humanos , Masculino , Mioblastos de Músculo Liso/citologia , Células-Tronco Pluripotentes/citologia
7.
Artigo em Inglês | MEDLINE | ID: mdl-27965937

RESUMO

Objective: The study was designed to evaluate the role of Human cytomegalovirus (HCMV) infection on homebox (HOX) gene expression and the effects of overexpression of HOX genes on proliferation and apoptosis of vascular smooth muscle cells (VSMCs). Methods: Viral infection was verified by observation of cytopathic effects through inverted microscopy, viral particles by electron microscopy and HCMV IE gene amplification by RT-PCR. cDNA profiling technology was used to screen expression of HOX genes after HCMV infection in VSMCs. Abnormal expression of Haematopoietically-expressed homeobox (HHEX) was selected to construct over-expressed vector and transfected into VSMCs. The effects of over expression of HHEX on cell proliferation and apoptosis of VSMCs were assayed by flow cytometry. Apoptosis and proliferation-associated genes were also assayed by RT-PCR. Results: Multiple HOX gene expression levels had obvious changes after HCMV infection, among which expression of HHEX gene increased obviously at 24, 48, and 72 h after infection. Over expression of HHEX can promote VSMCs proliferation by promoting G0/G1 phase cells into S phase and inhibit VSMCs apoptosis. HHEX inhibited the expression of apoptosis-associated caspase 2 and caspase3 and promoted the expression of cell cycle-related genes such as CDK2 and CDK6, CyclinB2 and CyclinD2. Conclusion: HHEX over expression induced by HCMV infection closely associated with vascular proliferative diseases.


Assuntos
Apoptose , Proliferação de Células , Citomegalovirus/patogenicidade , Proteínas de Homeodomínio/biossíntese , Interações Hospedeiro-Patógeno , Músculo Liso Vascular/patologia , Mioblastos de Músculo Liso/fisiologia , Fatores de Transcrição/biossíntese , Células Cultivadas , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Mioblastos de Músculo Liso/virologia , Fatores de Transcrição/genética
8.
Neuroreport ; 27(12): 875-82, 2016 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-27391329

RESUMO

The present research was designed to investigate whether endothelin-1 (ET-1) secretion can be induced by oxyhemoglobin and whether nuclear factor κB (NF-κB) is involved in the regulation of ET-1 transcription in cerebrovascular muscle cells. Cerebrovascular muscle cells isolated from a rabbit basilar artery were stimulated by oxyhemoglobin (OxyHb) and ET-1 production was increased significantly in the supernatant. Inhibition of NF-κB with pyrrolidine dithiocarbamate and small interfering RNA decreased the expression of ET-1. Nuclear translocation of NF-κB and the degradation of IkB-α was observed with the stimulation of OxyHb. The supernatant obtained from cerebrovascular muscle cells stimulated by OxyHb produced contractions in arterial rings and was blocked by the ET-1 receptor antagonist (BQ-123). The time course of the OxyHb-induced contractions of the basilar artery rings correlated with the time course of the OxyHb-induced ET-1 secretion. The contraction of the basilar artery rings induced by OxyHb was attenuated when the artery rings were preincubated with pyrrolidine dithiocarbamate and SN50 (20 and 10 µM, respectively). These results indicate that cerebrovascular muscle cells may be an important source of ET-1 production after subarachnoid hemorrhage. NF-κB was involved in the expression of ET-1 and the inhibition of the NF-κB pathway may be beneficial for the treatment of cerebral vasospasm.


Assuntos
Artéria Basilar/metabolismo , Endotelina-1/metabolismo , Mioblastos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Oxiemoglobinas/administração & dosagem , Animais , Artéria Basilar/fisiologia , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Masculino , Contração Muscular , RNA Mensageiro/metabolismo , Coelhos , Hemorragia Subaracnóidea/metabolismo
9.
Circ Res ; 116(8): 1392-412, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25858065

RESUMO

The vasculature plays an indispensible role in organ development and maintenance of tissue homeostasis, such that disturbances to it impact greatly on developmental and postnatal health. Although cell turnover in healthy blood vessels is low, it increases considerably under pathological conditions. The principle sources for this phenomenon have long been considered to be the recruitment of cells from the peripheral circulation and the re-entry of mature cells in the vessel wall back into cell cycle. However, recent discoveries have also uncovered the presence of a range of multipotent and lineage-restricted progenitor cells in the mural layers of postnatal blood vessels, possessing high proliferative capacity and potential to generate endothelial, smooth muscle, hematopoietic or mesenchymal cell progeny. In particular, the tunica adventitia has emerged as a progenitor-rich compartment with niche-like characteristics that support and regulate vascular wall progenitor cells. Preliminary data indicate the involvement of some of these vascular wall progenitor cells in vascular disease states, adding weight to the notion that the adventitia is integral to vascular wall pathogenesis, and raising potential implications for clinical therapies. This review discusses the current body of evidence for the existence of vascular wall progenitor cell subpopulations from development to adulthood and addresses the gains made and significant challenges that lie ahead in trying to accurately delineate their identities, origins, regulatory pathways, and relevance to normal vascular structure and function, as well as disease.


Assuntos
Doenças Cardiovasculares/patologia , Células Progenitoras Endoteliais/patologia , Músculo Liso Vascular/patologia , Mioblastos de Músculo Liso/patologia , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/cirurgia , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/transplante , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/transplante , Mioblastos de Músculo Liso/metabolismo , Mioblastos de Músculo Liso/transplante , Neovascularização Patológica , Neovascularização Fisiológica , Regeneração , Medicina Regenerativa/métodos , Nicho de Células-Tronco
10.
PLoS One ; 9(1): e86100, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475076

RESUMO

Interstitial cells of Cajal (ICC) are critical to gastrointestinal motility. The phenotypes of ICC progenitors have been observed in the mouse gut, but whether they exist in the human colon and what abnormal changes in their quantity and ultrastructure are present in Hirschsprung's disease (HSCR) colon remains uncertain. In this study, we collected the surgical resection of colons, both proximal and narrow segments, from HSCR patients and normal controls. First, we identified the progenitor of ICC in normal adult colon using immunofluorescent localization techniques with laser confocal microscopy. Next, the progenitors were sorted to observe their morphology. We further applied flow cytometry to examine the content of ICC progenitors in these fresh samples. The ultrastructural changes in the narrow and proximal parts of the HSCR colon were observed using transmission electron microscopy (TEM) and were compared with the normal adult colon. The presumed early progenitor (c-Kit(low)CD34(+)Igf1r(+)) and committed progenitor (c-Kit(+)CD34(+)Igf1r(+)) of ICC exist in adult normal colon as well as in the narrow and proximal parts of the HSCR colon. However, the proportions of mature, early and committed progenitors of ICC were dramatically reduced in the narrow segment of the HSCR colon. The proportions of mature and committed progenitors of ICC in the proximal segment of the HSCR colon were lower than in the adult normal colon. Ultrastructurally, ICC, enteric nerves, and smooth muscle in the narrow segment of the HSCR colon showed severe injury, including swollen vacuola or ted mitochondria, disappearance of mitochondrial cristae, dilated rough endoplasmic reticulum, vesiculation and degranulation, and disappearance of the caveolae on the ICC membrane surface. The contents of ICC and its progenitors in the narrow part of the HSCR colon were significantly decreased than those of adult colon, which may be associated with HSCR pathogenesis.


Assuntos
Doença de Hirschsprung/metabolismo , Células Intersticiais de Cajal/metabolismo , Mioblastos de Músculo Liso/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Células Cultivadas , Pré-Escolar , Feminino , Humanos , Imunofenotipagem , Lactente , Células Intersticiais de Cajal/citologia , Células Intersticiais de Cajal/ultraestrutura , Masculino , Pessoa de Meia-Idade , Mioblastos de Músculo Liso/citologia , Mioblastos de Músculo Liso/ultraestrutura , Fenótipo
11.
Kardiol Pol ; 71(10): 1048-58, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24197586

RESUMO

BACKGROUND: Modern therapies of post infarcted heart failure are focused on perfusion improvement of the injured myocardium. This effect can be achieved by, among other means, implanting stem cells which could be genetically modified with factors inducing the formation of new blood vessels in the post infarction scar area. Combined stem cell and gene therapy seems to be a promising strategy to heal an impaired myocardium. The creation of new blood vessels can be indirectly stimulated via factors inducing vascular endothelial growth factor synthesis, for example endothelial nitric oxide synthase (eNOS). The product of this enzyme, nitric oxide, is a molecule that can influence numerous physiological activities; it can contribute to vasodilation, stimulation of endothelial cell growth, prevention of platelet aggregation and leukocyte adhesion to the endothelium. AIM: To verify the pro-angiogenic and regenerative potential of human primary myoblasts and murine myoblast cell line C2C12 transiently transfected with eNOS gene. METHODS: Stem cells (either human or murine) were maintained in standard in vitro conditions. Next, both types of myoblasts were modified using electroporation and lipofection (human and murine cells), respectively. The efficacy of the transfection method was evaluated using flow cytometry. The concentration of eNOS protein was measured by ELISA immunoassay. The biological properties of modified cells were assessed using an MTT proliferation test and DAPI cell cycle analysis. To verify the influence of oxidative stress on myoblasts, cytometric tests using Annexin V and propidium iodide were applied. To check possible alterations in myogenic gene expression of stem cells transduced by genetic modification, the myogenic regulatory factors were evaluated by real-time PCR. The function of genetic modification was confirmed by a HUVEC capillary sprouting test using myoblasts supernatants. RESULTS: Electroporation turned out to be an efficient transfection method. High amounts of secreted protein were obtained (in the range 2,000 pg/mL) in both cell types studied. Moreover, the functionality of gene overexpression product was confirmed in capillary development assay. Human myoblasts did not exhibit any changes in cell cycle; however, eNOS transfected murine myoblasts revealed a statistically significant reduction in cell cycle ratio compared to controls (p < 0.001). In the case of myogenic gene expression, a decrease in Myogenin level was only detected in the human transfected myoblast population (p < 0.05). CONCLUSIONS: The results of our study may suggest that transplantation of myoblasts overexpressing eNOS could be promising for cell therapy in regenerating the post infarction heart.


Assuntos
Terapia Genética , Mioblastos Esqueléticos/transplante , Mioblastos de Músculo Liso/transplante , Infarto do Miocárdio/terapia , Óxido Nítrico Sintase Tipo III/genética , Células-Tronco/citologia , Animais , Apoptose/genética , Ciclo Celular/genética , Proliferação de Células , Células Cultivadas , Eletroporação , Células Endoteliais/citologia , Humanos , Camundongos , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Mioblastos de Músculo Liso/citologia , Mioblastos de Músculo Liso/metabolismo , Neovascularização Fisiológica/genética , Estresse Oxidativo/genética , Regeneração/genética , Transplante de Células-Tronco , Transfecção , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular
12.
J Surg Res ; 182(1): 40-8, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22925499

RESUMO

PURPOSE: Acellular porcine small intestinal submucosa (SIS) has been successfully used for esophagoplasty in dogs. However, this has not led to complete epithelialization and muscular regeneration. We undertook the present study to assess the effect of tissue-engineered esophagus generated by seeding bone marrow mesenchymal stem cells (BMSCs) onto an SIS scaffold (BMSCs-SIS) in a canine model. METHODS: We cultured, passaged, and measured autologous BMSCs and myoblasts with cell proliferation and immunohistochemical assays. We labeled the third passage of BMSCs with PKH-26, a fluorescent dye, before seeded it onto the SIS. We resected canine cervical esophagus to generate a defect 5 cm in length and 50% in circumference, which we repaired with BMSCs-SIS or SIS alone. RESULTS: Four weeks later, barium esophagram demonstrated that esophageal lumen surface of the patch graft was smoother in the BMSCs-SIS group compared with the SIS group. Histological examination suggested a strong similarity between BMSCs and esophageal myoblasts in terms of morphology and function. Although both BMSCs-SIS and SIS repaired the esophageal defects, we noted complete re-epithelialization with almost no inflammation only in the former group. By 12 wk after the surgery, we observed long bundles of skeletal muscles only in the BMSCs-SIS group, where the microvessel density was also much greater. CONCLUSIONS: Bone marrow mesenchymal stem cells on an SIS scaffold can promote re-epithelialization, revascularization, and muscular regeneration. This approach may provide an attractive option for esophageal regeneration.


Assuntos
Diferenciação Celular/fisiologia , Esôfago/citologia , Células-Tronco Mesenquimais/citologia , Modelos Animais , Engenharia Tecidual/métodos , Actinas/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Proliferação de Células , Células Cultivadas , Cães , Esôfago/fisiologia , Masculino , Células-Tronco Mesenquimais/fisiologia , Mioblastos de Músculo Liso/citologia , Mioblastos de Músculo Liso/metabolismo , Regeneração/fisiologia , Alicerces Teciduais
13.
Development ; 139(17): 3099-108, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22833126

RESUMO

Smooth muscle cells (SMCs) are a key component of many visceral organs, including the ureter, yet the molecular pathways that regulate their development from mesenchymal precursors are insufficiently understood. Here, we identified epithelial Wnt7b and Wnt9b as possible ligands of Fzd1-mediated ß-catenin (Ctnnb1)-dependent (canonical) Wnt signaling in the adjacent undifferentiated ureteric mesenchyme. Mice with a conditional deletion of Ctnnb1 in the ureteric mesenchyme exhibited hydroureter and hydronephrosis at newborn stages due to functional obstruction of the ureter. Histological analysis revealed that the layer of undifferentiated mesenchymal cells directly adjacent to the ureteric epithelium did not undergo characteristic cell shape changes, exhibited reduced proliferation and failed to differentiate into SMCs. Molecular markers for prospective SMCs were lost, whereas markers of the outer layer of the ureteric mesenchyme fated to become adventitial fibroblasts were expanded to the inner layer. Conditional misexpression of a stabilized form of Ctnnb1 in the prospective ureteric mesenchyme resulted in the formation of a large domain of cells that exhibited histological and molecular features of prospective SMCs and differentiated along this lineage. Our analysis suggests that Wnt signals from the ureteric epithelium pattern the ureteric mesenchyme in a radial fashion by suppressing adventitial fibroblast differentiation and initiating smooth muscle precursor development in the innermost layer of mesenchymal cells.


Assuntos
Hipoxantina Fosforribosiltransferase/genética , Mioblastos de Músculo Liso/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Ureter/embriologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Diferenciação Celular/fisiologia , Cruzamentos Genéticos , Fluorescência , Técnicas de Introdução de Genes , Hibridização In Situ , Camundongos , Mioblastos de Músculo Liso/metabolismo , Ureter/citologia , Ureter/metabolismo , beta Catenina/deficiência
14.
Differentiation ; 84(1): 41-53, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22652098

RESUMO

The importance of the epicardium covering the heart and the intrapericardial part of the great arteries has reached a new summit. It has evolved as a major cellular component with impact both in development, disease and more recently also repair potential. The role of the epicardium in development, its differentiation from a proepicardial organ at the venous pole (vPEO) and the differentiation capacities of the vPEO initiating cardiac epicardium (cEP) into epicardium derived cells (EPDCs) have been extensively described in recent reviews on growth and transcription factor pathways. In short, the epicardium is the source of the interstitial, the annulus fibrosus and the adventitial fibroblasts, and differentiates into the coronary arterial smooth muscle cells. Furthermore, EPDCs induce growth of the compact myocardium and differentiation of the Purkinje fibers. This review includes an arterial pole located PEO (aPEO) that provides the epicardium covering the intrapericardial great vessels. In avian and mouse models disturbance of epicardial outgrowth and maturation leads to a broad spectrum of cardiac anomalies with main focus on non-compaction of the myocardium, deficient annulus fibrosis, valve malformations and coronary artery abnormalities. The discovery that in disease both arterial and cardiac epicardium can again differentiate into EPDCs and thus reactivate its embryonic program and potential has highly broadened the scope of research interest. This reactivation is seen after myocardial infarction and also in aneurysm formation of the ascending aorta. Use of EPDCs for cell therapy show their positive function in paracrine mediated repair processes which can be additive when combined with the cardiac progenitor stem cells that probably share the same embryonic origin with EPDCs. Research into the many cell-autonomous and cell-cell-based capacities of the adult epicardium will open up new realistic therapeutic avenues.


Assuntos
Linhagem da Célula , Cardiopatias Congênitas/embriologia , Pericárdio/citologia , Pericárdio/embriologia , Animais , Diferenciação Celular , Transplante de Células , Embrião de Galinha , Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Fibroblastos/citologia , Cardiopatias Congênitas/terapia , Humanos , Camundongos , Mioblastos Cardíacos/citologia , Mioblastos de Músculo Liso/citologia , Miócitos Cardíacos/citologia , Miócitos de Músculo Liso/citologia , Pericárdio/fisiopatologia
15.
Dev Biol ; 367(2): 178-86, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22609551

RESUMO

MicroRNAs are potent modulators of cellular differentiation. miR-145 is expressed in, and promotes the differentiation of vascular and visceral smooth muscle cells (SMCs). Interestingly, we have observed that miR-145 also promotes differentiation of the gut epithelium in the developing zebrafish, a cell type where it is not expressed. Here we identify that a paracrine pathway involving the morphogens Sonic hedgehog (Shh) in epithelium and bone morphogenic protein 4 (Bmp4) in SMCs is modulated by miR-145. We show that expression of miR-145 in visceral SMCs normally represses the expression of the morphogen bmp4, as loss of miR-145 leads to upregulation of bmp4 in SMCs. We show that bmp4 in turn controls expression of Shh in the visceral epithelium. Conversely, in miR-145 morphants where bmp4 expression is increased, expression of sonic hedgehog a (shha) is strongly increased in gut epithelium. We show that expression of bmp4 is modulated by the miR-145 direct target gata6 but not a second potential direct target, klf5a. Thus although miR-145 is a tissue-restricted microRNA, it plays an essential role in promoting the patterning of both gut layers during gut development via a paracrine mechanism.


Assuntos
MicroRNAs/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Sequência de Bases , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Proliferação de Células , Sistema Digestório/embriologia , Sistema Digestório/metabolismo , Fatores de Transcrição GATA/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Morfolinos/genética , Músculo Liso/embriologia , Músculo Liso/metabolismo , Mioblastos de Músculo Liso/citologia , Mioblastos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Comunicação Parácrina , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
16.
Proc Natl Acad Sci U S A ; 109(18): 6993-8, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22509029

RESUMO

Vascular smooth muscle cells (VSMC) have been suggested to arise from various developmental sources during embryogenesis, depending on the vascular bed. However, evidence also points to a common subpopulation of vascular progenitor cells predisposed to VSMC fate in the embryo. In the present study, we use binary transgenic reporter mice to identify a Tie1(+)CD31(dim)vascular endothelial (VE)-cadherin(-)CD45(-) precursor that gives rise to VSMC in vivo in all vascular beds examined. This precursor does not represent a mature endothelial cell, because a VE-cadherin promoter-driven reporter shows no expression in VSMC during murine development. Blockade of Notch signaling in the Tie1(+) precursor cell, but not the VE-cadherin(+) endothelial cell, decreases VSMC investment of developing arteries, leading to localized hemorrhage in the embryo at the time of vascular maturation. However, Notch signaling is not required in the Tie1(+) precursor after establishment of a stable artery. Thus, Notch activity is required in the differentiation of a Tie1(+) local precursor to VSMC in a spatiotemporal fashion across all vascular beds.


Assuntos
Diferenciação Celular/fisiologia , Mioblastos de Músculo Liso/citologia , Mioblastos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Neovascularização Fisiológica , Receptores Notch/metabolismo , Animais , Antígenos CD/genética , Artérias/embriologia , Artérias/crescimento & desenvolvimento , Artérias/metabolismo , Sequência de Bases , Caderinas/deficiência , Caderinas/genética , Diferenciação Celular/genética , Primers do DNA/genética , Feminino , Antígenos Comuns de Leucócito/deficiência , Antígenos Comuns de Leucócito/genética , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Gravidez , Receptor de TIE-1/metabolismo , Receptores Notch/antagonistas & inibidores , Transdução de Sinais
18.
Arterioscler Thromb Vasc Biol ; 32(1): e1-11, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21979435

RESUMO

OBJECTIVE: Transforming growth factor-ß (TGF-ß) signaling is required for normal vascular development. We aimed to discover the role of TGF-ß signaling in embryonic smooth muscle cells (SMCs). METHODS AND RESULTS: We bred mice with smooth muscle (SM) 22α-Cre and Tgfbr2(flox) alleles to generate embryos in which the type II TGF-ß receptor (TGFBR2; required for TGF-ß signaling) was deleted in SMCs. Embryos were harvested between embryonic day (E) 9.5 and E18.5 and examined grossly, microscopically, and by histochemical and RNA analyses. SM22α-Cre(+/0) Tgfbr2(flox/flox) (knockout [KO]) embryos died before E15.5 with defects that included cardiac outflow tract abnormalities, persistence of the right dorsal aorta, and dilation of the distal aorta. Histological analyses suggested normal expression of SMC differentiation markers in KO aortas; however, RNA analyses showed that SMC differentiation markers were increased in KO cardiac outflow vessels but decreased in the descending aorta. KO aortas had only rare mature elastin deposits and contained abnormal aggregates of extracellular matrix proteins. Expression of several matrix proteins was significantly decreased in KO descending aortas but not in cardiac outflow vessels. CONCLUSIONS: TGF-ß signaling in SMCs controls differentiation, matrix synthesis, and vascular morphogenesis. Effects of TGF-ß on SMC gene expression appear to differ depending on the location of SMCs in the aorta.


Assuntos
Mioblastos de Músculo Liso/citologia , Mioblastos de Músculo Liso/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Aorta/anormalidades , Aorta/embriologia , Aorta/metabolismo , Vasos Sanguíneos/embriologia , Diferenciação Celular/fisiologia , Proteínas da Matriz Extracelular/biossíntese , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Desenvolvimento Muscular/fisiologia , Neovascularização Fisiológica , Gravidez , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Transdução de Sinais
19.
Arterioscler Thromb Vasc Biol ; 32(1): 42-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22034512

RESUMO

OBJECTIVE: The goal of this study was to use mice expressing human tissue factor pathway inhibitor (TFPI) on α-smooth muscle actin (α-SMA)(+) cells as recipients of allogeneic aortas to gain insights into the cellular mechanisms of intimal hyperplasia (IH). METHODS AND RESULTS: BALB/c aortas (H-2(d)) transplanted into α-TFPI-transgenic (Tg) mice (H-2(b)) regenerated a quiescent endothelium in contrast to progressive IH seen in C57BL/6 wild-type (WT) mice even though both developed aggressive anti-H-2(d) alloresponses, indicating similar vascular injuries. Adoptively transferred Tg CD34(+) (but not CD34(-)) cells inhibited IH in WT recipients, indicating the phenotype of α-TFPI-Tg mice was due to these cells. Compared with syngeneic controls, endogenous CD34(+) cells were mobilized in significant numbers after allogeneic transplantation, the majority showing sustained expression of tissue factor and protease-activated receptor-1 (PAR-1). In WT, most were CD45(+) myeloid progenitors coexpressing CD31, vascular endothelial growth factor receptor-2 and E-selectin; 10% of these cells coexpressed α-SMA and were recruited to the neointima. In contrast, the α-SMA(+) human TFPI(+) CD34(+) cells recruited in Tg recipients were from a CD45(-) lineage. WT CD34(+) cells incubated with a PAR-1 antagonist or taken from PAR-1-deficient mice inhibited IH as Tg cells did. CONCLUSIONS: Specific inhibition of thrombin generation or PAR-1 signaling on α-SMA(+) CD34(+) cells inhibits IH and promotes regenerative repair despite ongoing immune-mediated damage.


Assuntos
Actinas/metabolismo , Lesões das Artérias Carótidas/metabolismo , Mioblastos de Músculo Liso/metabolismo , Receptores de Trombina/antagonistas & inibidores , Transferência Adotiva , Animais , Antígenos CD34/metabolismo , Aorta/imunologia , Aorta/metabolismo , Aorta/patologia , Aorta/transplante , Lesões das Artérias Carótidas/imunologia , Lesões das Artérias Carótidas/patologia , Humanos , Lipoproteínas/genética , Lipoproteínas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mioblastos de Músculo Liso/imunologia , Mioblastos de Músculo Liso/patologia , Neointima/imunologia , Neointima/metabolismo , Neointima/patologia , Receptor PAR-1/antagonistas & inibidores , Receptor PAR-1/deficiência , Receptor PAR-1/genética , Receptores de Trombina/metabolismo , Transdução de Sinais , Cicatrização/fisiologia
20.
Genomics ; 98(6): 401-11, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21907276

RESUMO

Identifying gene regulatory elements and their target genes in human cells remains a significant challenge. Despite increasing evidence of physical interactions between distant regulatory elements and gene promoters in mammalian cells, many studies consider only promoter-proximal regulatory regions. We identify putative cis-regulatory modules (CRMs) in human skeletal muscle differentiation by combining myogenic TF binding data before and after differentiation with histone modification data in myoblasts. CRMs that are distant (>20 kb) from muscle gene promoters are common and are more likely than proximal promoter regions to show differentiation-specific changes in myogenic TF binding. We find that two of these distant CRMs, known to activate transcription in differentiating myoblasts, interact physically with gene promoters (PDLIM3 and ACTA1) during differentiation. Our results highlight the importance of considering distal CRMs in investigations of mammalian gene regulation and support the hypothesis that distant CRM-promoter looping contacts are a general mechanism of gene regulation.


Assuntos
Diferenciação Celular/genética , Elementos Facilitadores Genéticos , Músculo Esquelético/citologia , Mioblastos de Músculo Liso/citologia , Algoritmos , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Mioblastos de Músculo Liso/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica/genética , Análise de Sequência de DNA , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA