Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Orthop Surg Res ; 18(1): 876, 2023 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-37980502

RESUMO

BACKGROUND: Human bone marrow mesenchymal stem cells (hBMSCs) are a major source of osteoblast precursor cells and are directly involved in osteoporosis (OP) progression. Bromodomain-containing protein 4 (BRD4) is an important regulator for osteogenic differentiation. Therefore, its role and mechanism in osteogenic differentiation process deserve further investigation. METHODS: hBMSCs osteogenic differentiation was evaluated by flow cytometry, alkaline phosphatase assay and alizarin red staining. Western blot was used to test osteogenic differentiation-related proteins, BRD4 protein, WNT family members-4 (WNT4)/NF-κB-related proteins, and glycolysis-related proteins. Metabolomics techniques were used to detect metabolite changes and metabolic pathways. BRD4 and WNT4 mRNA levels were determined using quantitative real-time PCR. Dual-luciferase reporter assay and chromatin immunoprecipitation assay were performed to detect BRD4 and WNT4 interaction. Glycolysis ability was assessed by testing glucose uptake, lactic acid production, and ATP levels. RESULTS: After successful induction of osteogenic differentiation, the expression of BRD4 was increased significantly. BRD4 knockdown inhibited hBMSCs osteogenic differentiation. Metabolomics analysis showed that BRD4 expression was related to glucose metabolism in osteogenic differentiation. Moreover, BRD4 could directly bind to the promoter of the WNT4 gene. Further experiments confirmed that recombinant WNT4 reversed the inhibition effect of BRD4 knockdown on glycolysis, and NF-κB inhibitors (Bardoxolone Methyl) overturned the suppressive effect of BRD4 knockdown on hBMSCs osteogenic differentiation. CONCLUSION: BRD4 promoted hBMSCs osteogenic differentiation by inhibiting NF-κB pathway via enhancing WNT4 expression.


Assuntos
Células-Tronco Mesenquimais , MicroRNAs , Humanos , NF-kappa B/metabolismo , Osteogênese , Proteínas Nucleares/metabolismo , MicroRNAs/genética , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Células Cultivadas , Células da Medula Óssea/metabolismo , Proteína Wnt4/metabolismo , Proteína Wnt4/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Ciclo Celular
2.
Nature ; 586(7830): 606-611, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32814902

RESUMO

Islets derived from stem cells hold promise as a therapy for insulin-dependent diabetes, but there remain challenges towards achieving this goal1-6. Here we generate human islet-like organoids (HILOs) from induced pluripotent stem cells and show that non-canonical WNT4 signalling drives the metabolic maturation necessary for robust ex vivo glucose-stimulated insulin secretion. These functionally mature HILOs contain endocrine-like cell types that, upon transplantation, rapidly re-establish glucose homeostasis in diabetic NOD/SCID mice. Overexpression of the immune checkpoint protein programmed death-ligand 1 (PD-L1) protected HILO xenografts such that they were able to restore glucose homeostasis in immune-competent diabetic mice for 50 days. Furthermore, ex vivo stimulation with interferon-γ induced endogenous PD-L1 expression and restricted T cell activation and graft rejection. The generation of glucose-responsive islet-like organoids that are able to avoid immune detection provides a promising alternative to cadaveric and device-dependent therapies in the treatment of diabetes.


Assuntos
Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Evasão da Resposta Imune , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/imunologia , Organoides/citologia , Organoides/imunologia , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linhagem Celular , Epigênese Genética , Feminino , Glucose/metabolismo , Rejeição de Enxerto , Xenoenxertos , Homeostase , Humanos , Tolerância Imunológica , Secreção de Insulina , Transplante das Ilhotas Pancreáticas , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Organoides/transplante , Linfócitos T/citologia , Linfócitos T/imunologia , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt4/metabolismo , Proteína Wnt4/farmacologia
3.
Biochem Biophys Res Commun ; 479(4): 793-799, 2016 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-27687546

RESUMO

The Wnt signalling pathway in beta-cells has been linked to the development of type 2 diabetes. Investigating the impact of a non-canonical Wnt ligand, Wnt4, on beta-cell function we found that in INS-1 cells, Wnt4 was able to completely block Wnt3a stimulated cell growth and insulin secretion. However, despite high levels of Wnt4 protein being detected in INS-1 cells, reducing the expression of Wnt4 had no impact on cell growth or Wnt3a signalling. As such, the role of the endogenously expressed Wnt4 in beta-cells is unclear, but the data showing that Wnt4 can act as a negative regulator of canonical Wnt signalling in beta-cells suggests that this pathway could be a potential target for modulating beta-cell function.


Assuntos
Células Secretoras de Insulina/metabolismo , Proteína Wnt3A/metabolismo , Proteína Wnt4/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Glucose/metabolismo , Glucose/farmacologia , Humanos , Imuno-Histoquímica , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Ratos , Via de Sinalização Wnt , Proteína Wnt3A/antagonistas & inibidores , Proteína Wnt3A/farmacologia , Proteína Wnt4/genética , Proteína Wnt4/farmacologia , beta Catenina/genética , beta Catenina/metabolismo
4.
PLoS One ; 8(6): e64843, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23755150

RESUMO

Embryonic stem cells and induced pluripotent stem cells have the ability to differentiate into various organs and tissues, and are regarded as new tools for the elucidation of disease mechanisms as well as sources for regenerative therapies. However, a method of inducing organ-specific cells from pluripotent stem cells is urgently needed. Although many scientists have been developing methods to induce various organ-specific cells from pluripotent stem cells, renal lineage cells have yet to be induced in vitro because of the complexity of kidney structures and the diversity of kidney-component cells. Here, we describe a method of inducing renal tubular cells from mouse embryonic stem cells via the cell purification of kidney specific protein (KSP)-positive cells using an anti-KSP antibody. The global gene expression profiles of KSP-positive cells derived from ES cells exhibited characteristics similar to those of cells in the developing kidney, and KSP-positive cells had the capacity to form tubular structures resembling renal tubular cells when grown in a 3D culture in Matrigel. Moreover, our results indicated that KSP-positive cells acquired the characteristics of each segment of renal tubular cells through tubular formation when stimulated with Wnt4. This method is an important step toward kidney disease research using pluripotent stem cells, and the development of kidney regeneration therapies.


Assuntos
Caderinas/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Túbulos Renais/citologia , Proteínas/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Caderinas/química , Diferenciação Celular/efeitos dos fármacos , Colágeno/farmacologia , Combinação de Medicamentos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Espaço Extracelular/química , Fator de Crescimento de Hepatócito/farmacologia , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/farmacologia , Túbulos Renais/embriologia , Túbulos Renais/ultraestrutura , Laminina/farmacologia , Camundongos , Células NIH 3T3 , Especificidade de Órgãos/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteínas/química , Proteoglicanas/farmacologia , Proteína Wnt4/farmacologia
5.
J Am Soc Nephrol ; 24(9): 1399-412, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23766539

RESUMO

Injury to the adult kidney induces a number of developmental genes thought to regulate repair, including Wnt4. During kidney development, early nephron precursors and medullary stroma both express Wnt4, where it regulates epithelialization and controls smooth muscle fate, respectively. Expression patterns and roles for Wnt4 in the adult kidney, however, remain unclear. In this study, we used reporters, lineage analysis, and conditional knockout or activation of the Wnt/ß-catenin pathway to investigate Wnt4 in the adult kidney. Proliferating, medullary, interstitial myofibroblasts strongly expressed Wnt4 during renal fibrosis, whereas tubule epithelia, except for the collecting duct, did not. Exogenous Wnt4 drove myofibroblast differentiation of a pericyte-like cell line, suggesting that Wnt4 might regulate pericyte-to-myofibroblast transition through autocrine signaling. However, conditional deletion of Wnt4 in interstitial cells did not reduce myofibroblast proliferation, cell number, or myofibroblast gene expression during fibrosis. Because the injured kidney expresses multiple Wnt ligands that might compensate for the absence of Wnt4, we generated a mouse model with constitutive activation of canonical Wnt/ß-catenin signaling in interstitial pericytes and fibroblasts. Kidneys from these mice exhibited spontaneous myofibroblast differentiation in the absence of injury. Taken together, Wnt4 expression in renal fibrosis defines a population of proliferating medullary myofibroblasts. Although Wnt4 may be dispensable for myofibroblast transformation, canonical Wnt signaling through ß-catenin stabilization is sufficient to drive spontaneous myofibroblast differentiation in interstitial pericytes and fibroblasts, emphasizing the importance of this pathway in renal fibrosis.


Assuntos
Medula Renal/irrigação sanguínea , Miofibroblastos/fisiologia , Traumatismo por Reperfusão/fisiopatologia , Transdução de Sinais/fisiologia , Obstrução Ureteral/fisiopatologia , Proteína Wnt4/fisiologia , beta Catenina/fisiologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Deleção de Genes , Técnicas In Vitro , Medula Renal/patologia , Medula Renal/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miofibroblastos/patologia , Traumatismo por Reperfusão/patologia , Obstrução Ureteral/patologia , Urotélio/patologia , Urotélio/fisiopatologia , Proteína Wnt4/genética , Proteína Wnt4/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA