Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Cell Physiol Biochem ; 47(5): 1785-1799, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29953975

RESUMO

Backgroud/Aims: Mesenchymal stromal cells (MSCs) are a major component of the tumor microenvironment (TME). Several studies focusing on tumor-derived MSCs have demonstrated that they exhibit a strong ability to promote the tumor epithelial-mesenchymal transition (EMT). However, the factors mediating these effects are poorly understood. METHODS: Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and immunohistochemistry assays were used to detect the expression of Gremlin1 (GREM1) in human esophageal squamous cell carcinoma (ESCC) tissues. ShRNA silencing, flow cytometry, cell counting kit (CCK8) assay, invasion assay, western blot were used to detect the effect of GREM1 in ECa109, TE-1 cell lines and xenograft tumor models. RESULTS: In the current study, we found that the GREM1 was overexpressed in human ESCC tissues. The conditioned medium from mesenchymal stromal cells (MSCs-CM) enhanced the malignancy of xenograft esophageal tumors in vivo, as well as the cell proliferation, viability and invasion of the esophageal carcinoma cell lines ECa109 and TE-1 in vitro. Furthermore, the shRNA silencing of GREM1 in MSCs (shGREM1-MSCs) reversed the increased malignancy of the esophageal tumor in vivo, while the conditioned medium from shGREM1-MSCs (shGREM1-MSCs-CM) affected the cell cycle and cell invasion in vitro. These processes were accompanied by the EMT in the ECa109 and TE-1 cell lines with an alteration in the expression levels of mesenchymal and epithelial markers. Furthermore, the TGF-ß/BMP (transforming growth factor-beta/bone morphogenetic protein) signaling pathway participated in the shGREM1-MSCs-CM-induced anti-tumor effect on enhanced esophageal malignancy induced by MSCs-CM treatment. CONCLUSIONS: Taken together, our study suggested that GREM1 delivered by MSCs promoted EMT in ESCC in vitro and in vivo, which is partly through TGF-ß/BMP signaling pathway. The results provide experimental evidence to a potential therapeutic target in the treatment of esophageal cancer.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Transição Epitelial-Mesenquimal , Neoplasias Esofágicas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas de Neoplasias/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Neoplasias Esofágicas/patologia , Humanos , Metaloproteinases da Matriz Secretadas/metabolismo , Células-Tronco Mesenquimais/patologia , Fator de Crescimento Transformador beta/metabolismo
2.
J Cell Physiol ; 232(5): 996-1007, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27579673

RESUMO

Mesenchymal stem cells (MSCs) are a promising cell resource for the treatment of ischemic diseases, partially through paracrine effects. One of the major obstacles of MSC treatment is the poor survival rate and low efficiency of transplanted stem cells due to ischemic or inflammatory environments. Gremlin1 (GREM1), a regulator of growth, differentiation and development, has been identified as a novel proangiogenic factor. However, the role and mechanism of GREM1 in MSCs remains unclear. Therefore, we assessed the putative beneficial effects of GREM1 on MSC-based therapy for hindlimb ischemia. The lentiviral vector, EF1a-GREM1, was constructed using the Multisite Gateway System and used to transduce MSCs. In vitro studies demonstrated increased survival of GREM1-MSCs exposed to H2 O2 , which is consistent with the activation of caspase-3. Conditional medium from GREM1-MSCs (GREM1-MSC-CM) increased the anti-apoptotic effects of human umbilical vein endothelial cells (HUVECs), and this effect was attenuated by treatment with the PI3K/Akt pathway inhibitor LY294002. MSCs modified with GREM1 could significantly increase blood perfusion of the ischemic hindlimb in vivo in a mouse model, which was correlated to improved MSC survival. This study demonstrates that overexpression of GREM1 in MSCs have greater therapeutic effects against ischemia compared with wild-type MSCs by enhancing the survival of MSCs and ECs, which may provide new tools for studies investigating the treatment of ischemic diseases. J. Cell. Physiol. 232: 996-1007, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Isquemia/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Apoptose , Arteríolas/patologia , Capilares/patologia , Sobrevivência Celular , Embrião de Galinha , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Isquemia/patologia , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neovascularização Fisiológica , Estresse Oxidativo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fluxo Sanguíneo Regional , Transdução de Sinais , Doadores de Tecidos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
Mol Ther ; 24(10): 1860-1872, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27397633

RESUMO

Mesenchymal stromal cells (MSCs) have shown great potential for treating inflammatory bowel disease, which is ameliorated through paracrine cross talk between MSCs and T-cells. Members of the insulin-like growth factor binding protein (IGFBP) family have important immunomodulatory functions in MSCs, but the underlying mechanisms behind these functions have not yet been clearly elucidated. In this study, we investigate whether MSC-produced IGFBP7 is involved in immune modulation using a mouse experimental colitis model. Gene expression profiling revealed that IGFBP7 was highly expressed in MSCs. Consistent with this findings, IGFBP7 knockdown in MSCs significantly decreased their immunomodulatory properties, decreasing the antiproliferative functions of MSCs against T-cells, while also having an effect on the proinflammatory cytokine production of the T-cells. Furthermore, in the mouse experimental colitis model, MSC-derived IGFBP7 ameliorated the clinical and histopathological severity of induced colonic inflammation and also restored the injured gastrointestinal mucosal tissues. In conclusion, IGFBP7 contributes significantly to MSC-mediated immune modulation, as is shown by the ability of IGFBP7 knockdown in MSCs to restore proliferation and cytokine production in T-cells. These results suggest that IGFBP7 may act as a novel MSC-secreted immunomodulatory factor.


Assuntos
Colite/terapia , Fatores Imunológicos/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Colite/induzido quimicamente , Colite/metabolismo , Modelos Animais de Doenças , Células-Tronco Mesenquimais/metabolismo , Camundongos , Regulação para Cima
4.
J Altern Complement Med ; 25(4): 435-440, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30431316

RESUMO

OBJECTIVE: To explore the association between green tea consumption and coronary arterial disease (CAD) in the Chinese population of Guangzhou. Design, location, subjects: A retrospective study at the Sun Yat-sen Memorial Hospital in Guangzhou, China. Consecutive patients were enrolled between January 2013 and August 2014. A total of 539 patients were included. Two hundred sixty-seven of them are CAD patients and 272 of them are non-CAD patients. The CAD patients were diagnosed according to international diagnostic criteria. INTERVENTIONS: Using data from the questionnaires and clinical laboratories, we attempted to elucidate the association between green tea and CAD. OUTCOME MEASURES: Baseline characteristics of study population, CAD-related biomarkers, amount, frequency and duration of green tea consumption, and CAD risk analysis. RESULTS: The results showed that among males, those who drank green tea did not have a reduced risk of CAD (odds ratio; OR = 1.58, 95% CI: 0.96-2.59, p > 0.05). However, women in the study who drank green tea had a reduced risk of CAD (OR = 0.13, 95% CI: 0.07-0.23, p < 0.01). The females who consumed ≤1 cup/day green tea had lower CAD risk (OR = 0.12, 95% CI: 0.07-0.23, p < 0.01). The frequency of 3-5 days/week (OR = 0.14, 95% CI: 0.07-0.29, p < 0.01) and >5 days/week (OR = 0.24, 95% CI: 0.08-0.69, p < 0.01) were both beneficial in preventing CAD. Those who had been drinking green tea for 0-10 years (OR = 0.11, 95% CI: 0.04-0.30), 10-20 years (OR = 0.22, 95% CI: 0.11-0.46), or >20 years (OR = 0.37, 95% CI: 0.12-0.96) had a reduced risk of CAD. CONCLUSIONS: Through the analysis of green tea consumption and CAD-related biomarkers, we concluded that a small amount of high-frequency green tea consumption was associated with a reduced risk of CAD in female populations in Guangzhou, China, and the association might be partly due to altered CAD-related biomarkers.


Assuntos
Doença da Artéria Coronariana/tratamento farmacológico , Doença da Artéria Coronariana/epidemiologia , Fitoterapia/estatística & dados numéricos , Chá , Idoso , China/epidemiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
5.
Stem Cell Res Ther ; 9(1): 51, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29482621

RESUMO

BACKGROUND: The LIM-homeobox transcription factor islet-1 (ISL1) has been proposed as a marker for cardiovascular progenitor cells. This study investigated whether forced expression of ISL1 in human mesenchymal stem cells (hMSCs) improves myocardial infarction (MI) treatment outcomes. METHODS: The lentiviral vector containing the human elongation factor 1α promoter, which drives the expression of ISL1 (EF1α-ISL1), was constructed using the Multisite Gateway System and used to transduce hMSCs. Flow cytometry, immunofluorescence, Western blotting, TUNEL assay, and RNA sequencing were performed to evaluate the function of ISL1-overexpressing hMSCs (ISL1-hMSCs). RESULTS: The in vivo results showed that transplantation of ISL1-hMSCs improved cardiac function in a rat model of MI. Left ventricle ejection fraction and fractional shortening were greater in post-MI hearts after 4 weeks of treatment with ISL1-hMSCs compared with control hMSCs or phosphate-buffered saline. We also found that ISL1 overexpression increased angiogenesis and decreased apoptosis and inflammation. The greater potential of ISL1-hMSCs may be attributable to an increased number of surviving cells after transplantation. Conditioned medium from ISL1-hMSCs decreased the apoptotic effect of H2O2 on the cardiomyocyte cell line H9c2. To clarify the molecular basis of this finding, we employed RNA sequencing to compare the apoptotic-related gene expression profiles of control hMSCs and ISL1-hMSCs. The results showed that insulin-like growth factor binding protein 3 (IGFBP3) was the only gene in ISL1-hMSCs with a RPKM value higher than 100 and that the difference fold-change between ISL1-hMSCs and control hMSCs was greater than 3, suggesting that IGFBP3 might play an important role in the anti-apoptosis effect of ISL1-hMSCs through paracrine effects. Furthermore, the expression of IGFBP3 in the conditioned medium from ISL1-hMSCs was almost fourfold greater than that in conditioned medium from control hMSCs. Moreover, the IGFBP3 neutralization antibody reversed the apoptotic effect of ISL1-hMSCs-CM. CONCLUSIONS: These results suggest that overexpression of ISL1 in hMSCs promotes cell survival in a model of MI and enhances their paracrine function to protect cardiomyocytes, which may be mediated through IGFBP3. ISL1 overexpression in hMSCs may represent a novel strategy for enhancing the effectiveness of stem cell therapy after MI.


Assuntos
Proteínas com Homeodomínio LIM/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/terapia , Fatores de Transcrição/genética , Animais , Células Cultivadas , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Células-Tronco Mesenquimais/citologia , Miócitos Cardíacos/metabolismo , Comunicação Parácrina , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA