Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
World J Gastroenterol ; 30(32): 3766-3782, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39221071

RESUMO

BACKGROUND: The efficacy of mesenchymal stem cells (MSCs) in treating liver fibrosis has been demonstrated in several clinical studies. However, their low survival and liver implantation rates remain problematic. In recent years, a large number of studies in animal models of liver fibrosis have shown that MSCs combined with drugs can improve the efficacy of MSCs in the treatment of liver fibrosis alone and inhibit its progression to end-stage liver disease. This has inspired new ways of thinking about treating liver fibrosis. AIM: To investigate the effectiveness and mechanisms of MSCs combined with drugs in treating liver fibrosis. METHODS: Data sources included four electronic databases and were constructed until January 2024. The subjects, interventions, comparators, outcomes, and study design principle were used to screen the literature, and the quality of the literature was evaluated to assess the risk of bias. Relevant randomised controlled trials were selected, and the final 13 studies were included in the final study. RESULTS: A total of 13 studies were included after screening. Pooled analysis showed that MSCs combined with drug therapy significantly improved liver function, promoted the repair of damaged liver tissues, reduced the level of liver fibrosis-related indexes, and effectively ameliorated hepatic fibrosis by modulating the hepatic inflammatory microenvironment, promoting the homing of MSCs, and regulating the relevant signaling pathways, and the treatment efficacy was superior to MSCs alone. However, the combined treatment statistics showed no ame-lioration in serum albumin levels (standardized mean difference = 0.77, 95% confidence interval: -0.13 to 1.68, P = 0.09). CONCLUSION: In conclusion, MSCs combined with drugs for treating liver fibrosis effectively make up for the shortcomings of MSCs in their therapeutic effects. However, due to the different drugs, the treatment mechanism and effect also differ. Therefore, more randomized controlled trials are needed to compare the therapeutic efficacy of different drugs in combination with MSCs, aiming to select the "best companion" of MSCs in treating hepatic fibrosis.


Assuntos
Cirrose Hepática , Transplante de Células-Tronco Mesenquimais , Animais , Humanos , Terapia Combinada/métodos , Modelos Animais de Doenças , Progressão da Doença , Fígado/patologia , Fígado/efeitos dos fármacos , Cirrose Hepática/patologia , Cirrose Hepática/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
2.
J Biol Chem ; 287(4): 2364-77, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22123824

RESUMO

Previous studies in our laboratory have shown that mixed lineage kinase 3 (MLK3) can be activated following global ischemia. In addition, other laboratories have reported that the activation of MLK3 may be linked to the accumulation of free radicals. However, the mechanism of MLK3 activation remains incompletely understood. We report here that MLK3, overexpressed in HEK293 cells, is S-nitrosylated (forming SNO-MLK3) via a reaction with S-nitrosoglutathione, an exogenous nitric oxide (NO) donor, at one critical cysteine residue (Cys-688). We further show that the S-nitrosylation of MLK3 contributes to its dimerization and activation. We also investigated whether the activation of MLK3 is associated with S-nitrosylation following rat brain ischemia/reperfusion. Our results show that the administration of 7-nitroindazole, an inhibitor of neuronal NO synthase (nNOS), or nNOS antisense oligodeoxynucleotides diminished the S-nitrosylation of MLK3 and inhibited its activation induced by cerebral ischemia/reperfusion. In contrast, 2-amino-5,6-dihydro-6-methyl-4H-1,3-thiazine (an inhibitor of inducible NO synthase) or nNOS missense oligodeoxynucleotides did not affect the S-nitrosylation of MLK3. In addition, treatment with sodium nitroprusside (an exogenous NO donor) and S-nitrosoglutathione or MK801, an antagonist of the N-methyl-D-aspartate receptor, also diminished the S-nitrosylation and activation of MLK3 induced by cerebral ischemia/reperfusion. The activation of MLK3 facilitated its downstream protein kinase kinase 4/7 (MKK4/7)-JNK signaling module and both nuclear and non-nuclear apoptosis pathways. These data suggest that the activation of MLK3 during the early stages of ischemia/reperfusion is modulated by S-nitrosylation and provides a potential new approach for stroke therapy whereby the post-translational modification machinery is targeted.


Assuntos
Isquemia Encefálica/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Multimerização Proteica , Processamento de Proteína Pós-Traducional , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Maleato de Dizocilpina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/genética , Masculino , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , S-Nitrosoglutationa/metabolismo , Tiazinas/farmacologia , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
3.
Curr Neurovasc Res ; 18(1): 20-77, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33745435

RESUMO

BACKGROUND: Previous studies have reported that mesenchymal stem cell (MSC)- derived exosomes can protect primary rat brain microvascular endothelial cells (BMECs) against oxygen-glucose deprivation and reoxygenation (OGD/R)-induced injury. OBJECTIVE: The aim was to identify the key factors mediating the protective effects of MSC-derived exosomes. METHODS: Primary rat BMECs were either pretreated or not pretreated with MSC-derived exosomes before exposure to OGD/R. Naïve cells were used as a control. After performing small RNA deep sequencing, quantitative reverse transcription polymerase chain reaction was performed to validate microRNA (miRNA) expression. The effects of rno-miR-666-3p on cell viability, apoptosis, and inflammation in OGD/R-exposed cells were assessed by performing the Cell Counting Kit 8 assay, flow cytometry, and enzyme-linked immunosorbent assay, respectively. Moreover, the role of rno-miR-666-3p in regulating gene expression in OGD/R-exposed cells was studied using mRNA deep sequencing. Lastly, to evaluate whether mitogen-activated protein kinase 1 (MAPK1) was the target of rno-miR-666-3p, western blotting and the dual-luciferase assay were performed. RESULTS: MSC-derived exosomes altered the miRNA expression patterns in OGD/R-exposed BMECs. In particular, the expression levels of rno-miR-666-3p, rno-miR-92a-2-5p, and rnomiR- 219a-2-3p decreased in OGD/R-exposed cells compared with those in the control; however, MSC-derived exosomes restored the expression levels of these miRNAs under OGD/R conditions. rno-miR-666-3p overexpression enhanced cell viability and alleviated the apoptosis of OGD/R-exposed cells. Moreover, rno-miR-666-3p suppressed OGD/R-induced inflammation. mRNA deep sequencing revealed that rno-miR-666-3p is closely associated with the MAPK signaling pathway. Western blotting and the dual-luciferase assay confirmed that MAPK1 is the target of rnomiR- 666-3p. CONCLUSION: MSC-derived exosomes restore rno-miR-666-3p expression in OGD/R-exposed BMECs. Moreover, this specific miRNA exerts protective effects against OGD/R by suppressing the MAPK signaling pathway.


Assuntos
Encéfalo/metabolismo , Sobrevivência Celular/fisiologia , Células Endoteliais/metabolismo , Exossomos/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Animais , Hipóxia Celular/fisiologia , Glucose/metabolismo , Oxigênio/metabolismo , Ratos
4.
Curr Neurovasc Res ; 17(2): 155-163, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32056526

RESUMO

OBJECTIVE: The effects of mesenchymal stem cell (MSC)-derived exosomes on brain microvascular endothelial cells under oxygen-glucose deprivation (OGD), which mimic cells in deep hypothermic circulatory arrest (DHCA) in vitro, are yet to be studied. METHODS: MSCs were co-cultured with primary rat brain endothelial cells, which were then exposed to OGD. Cell viability, apoptosis, the inflammatory factors (IL-1ß, IL-6, and TNF-α), and the activation of inflammation-associated TLR4-mediated pyroptosis and the NF-κB signaling pathway were determined. Furthermore, exosomes derived from MSCs were isolated and incubated with endothelial cells to investigate whether the effect of MSCs is associated with MSCderived exosomes. Apoptosis, cell viability, and the inflammatory response were also analyzed in OGD-induced endothelial cells incubated with MSC-derived exosomes. RESULTS: OGD treatment promoted endothelial cell apoptosis, induced the release of inflammatory factors IL-1ß, IL-6, and TNF-α, and inhibited cell viability. Western blot analysis showed that OGD treatment-induced TLR4, and NF-κB p65 subunit phosphorylation and caspase-1 upregulation, while co-culture with MSCs could reduce the effect of OGD treatment on endothelial cells. As expected, the effect of MSC-derived exosomes on OGD-treated endothelial cells was similar to that of MSCs. MSC-derived exosomes alleviated the OGD-induced decrease in the viability of endothelial cells, and increased levels of apoptosis, inflammatory factors, and the activation of inflammatory and inflammatory focal pathways. CONCLUSION: Both MSCs and MSC-derived exosomes attenuated OGD-induced rat primary brain endothelial cell injury. These findings suggest that MSC-derived exosomes mediate at least some of the protective effects of MSCs on endothelial cells.


Assuntos
Encéfalo/metabolismo , Hipóxia Celular/fisiologia , Células Endoteliais/metabolismo , Exossomos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Apoptose/fisiologia , Encéfalo/citologia , Sobrevivência Celular/fisiologia , Técnicas de Cocultura , Citocinas/metabolismo , Células Endoteliais/citologia , Glucose/metabolismo , Células-Tronco Mesenquimais/citologia , NF-kappa B/metabolismo , Oxigênio/metabolismo , Ratos , Transdução de Sinais/fisiologia
5.
Biomed Mater Eng ; 28(s1): S113-S119, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28372286

RESUMO

Liver transplantation is the definitive treatment for patients with end-stage liver diseases (ESLD). However, it is hampered by shortage of liver donor. Liver tissue engineering, aiming at fabricating new livers in vitro, provides a potential resolution for donor shortage. Three elements need to be considered in liver tissue engineering: seeding cell resources, scaffolds and bioreactors. Studies have shown potential cell sources as hepatocytes, hepatic cell line, mesenchymal stem cells and others. They need scaffolds with perfect biocompatiblity, suitable micro-structure and appropriate degradation rate, which are essential charateristics for cell attachment, proliferation and secretion in forming extracellular matrix. The most promising scaffolds in research include decellularized whole liver, collagens and biocompatible plastic. The development and function of cells in scaffold need a microenvironment which can provide them with oxygen, nutrition, growth factors, et al. Bioreactor is expected to fulfill these requirements by mimicking the living condition in vivo. Although there is great progress in these three domains, a large gap stays still between their researches and applications. Herein, we summarized the recent development in these three major fields which are indispensable in liver tissue engineering.


Assuntos
Materiais Biocompatíveis/química , Hepatócitos/citologia , Fígado/citologia , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Reatores Biológicos , Humanos , Fígado/crescimento & desenvolvimento , Fígado Artificial
6.
Brain Res ; 1634: 140-149, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26794251

RESUMO

Although studies have shown that cerebral ischemic preconditioning (IPC) can ameliorate ischemia/reperfusion (I/R) induced brain damage, but its precise mechanisms remain unknown. Therefore, the aim of this study was to investigate the neuroprotective mechanisms of IPC against ischemic brain damage induced by cerebral I/R and to explore whether the Calcium/calmodulin-dependent protein kinase II (CaMKII)-mediated up-regulation of nNOS ser847-phosphorylation signaling pathway contributed to the protection provided by IPC. Transient global brain ischemia was induced by 4-vessel occlusion in adult male Sprague-Dawley rats. The rats were pretreated with 3 min of IPC alone or KN62 (selective antagonist of CaMKII) treatment before IPC, after reperfusion for 3 days, 6 min ischemia was induced. Cresyl violet staining was used to examine the survival of hippocampal CA1 pyramidal neurons. Immunoblotting was performed to measure the phosphorylation of CaMKII, nNOS, c-Jun and the expression of FasL. Immunoprecipitation was used to examine the binding between PSD95 and nNOS. The results showed that IPC could significantly protect neurons against cerebral I/R injury, furthermore, the combination of PSD95 and nNOS was increased, coinstantaneously the phosphorylation of CaMKII and nNOS (ser847) were up-regulated, however the activation of c-Jun and FasL were reduced. Conversely, KN62 treatment before IPC reversed all these effects of IPC. Taken together, the results suggest that IPC could diminish ischemic brain injury through CaMKII-mediated up-regulation of nNOS ser847-phosphorylation signaling pathway.


Assuntos
Isquemia Encefálica/metabolismo , Região CA1 Hipocampal/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Precondicionamento Isquêmico , Óxido Nítrico Sintase Tipo I/metabolismo , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Região CA1 Hipocampal/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína 4 Homóloga a Disks-Large , Proteína Ligante Fas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Stem Cell Res Ther ; 6: 45, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25889932

RESUMO

INTRODUCTION: Bone marrow mesenchymal stem cells (BMSCs) have been studied extensively because of their potential use in clinical therapy, regenerative medicine, and tissue engineering. However, their application in tumor therapy remains yet in preclinical stage because of the distinct results from different researches and vagueness of their functional mechanism. In this study, the influence of BMSCs on tumor growth was observed and the potential mechanism was investigated. METHOD: Two animal models, H22 ascitogenous hepatoma in BALb/c mouse and B16-F10 pulmonary metastatic melanoma in C57 mouse, were adopted in experience in vivo and treated with BMSCs by intravenous injection. The percentage of Gr-1+CD11b+ myeloid-derived suppressor cells (MDSCs) and IFN-γ+ T cells were observed in peripheral blood (PB) and bone marrow (BM) by Flow Cytometry. BMSCs were co-cultured in vitro with tumor cells and MDSCs in a tumor conditioned medium separately in order to illustrate the mechanism. RESULTS: Our results demonstrated that BMSCs treatment caused a delayed tumor growth and a prolonged survival in both tumor models, the homing fraction of BMSCs in BM was 2% - 5% in 24-72 hours after transfusion and the percentage of Gr-1+CD11b+ MDSCs was downregulated in peripheral blood and BM. Meanwhile, IFN-γ+ T lymphocytes in PB increased. In vitro co-culture showed that BMSCs inhibited the induction and proliferation of MDSCs in tumor conditioned medium, whereas they didn't affect the proliferation of B16-F10 and H22 cells by in vitro co-culture. Both in vivo and in vitro results showed that BMSCs have a systemic suppressive effect on MDSCs. CONCLUSION: Our data suggest that BMSCs has suppressive effect on tumor and is feasible to be applied in cancer treatment. BMSCs inhibiting MDSCs induction and proliferation is likely one of the mechanism.


Assuntos
Transplante de Medula Óssea , Terapia Baseada em Transplante de Células e Tecidos/métodos , Melanoma Experimental/terapia , Transplante de Células-Tronco Mesenquimais , Linfócitos T/imunologia , Células 3T3 , Animais , Antineoplásicos , Células da Medula Óssea/fisiologia , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Interferon gama/metabolismo , Contagem de Linfócitos , Melanoma Experimental/patologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
8.
Biomed Mater Eng ; 25(1 Suppl): 137-43, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25538064

RESUMO

The shortage of organ resource has been limiting the application of liver transplantation. Bioartificial liver construction is increasingly focused as a replacement treatment. To product a bioartificial liver, three elements must be considered: seeding cells, scaffold and bioreactor. Recent studies have shown that several methods can successfully differentiate MSC (mesenchymal stem cells) derived from Wharton's jelly into hepatocyte, such as stimulating MSC by cytokines and growth factors, direct and indirect co-culture MSC with hepatocytes, or promote MSC differentiation by 3-dimensional matrix. In some cases, differentiation of MSC into hepatocytes can also be an alternative approach for whole organ transplantation in treatment of acute and chronic liver diseases. In this review, the characterization of MSC from Wharton's jelly, their potential of application in liver tissue engineering on base of decellularized scaffold, their status of banking and their preclinical work performed will be discussed.


Assuntos
Fígado Artificial , Células-Tronco Mesenquimais/citologia , Técnicas de Cultura de Órgãos/instrumentação , Engenharia Tecidual/instrumentação , Alicerces Teciduais , Geleia de Wharton/citologia , Reatores Biológicos , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Transplante de Células-Tronco Mesenquimais/instrumentação , Células-Tronco Mesenquimais/fisiologia , Desenho de Prótese
9.
Biomed Mater Eng ; 25(1 Suppl): 159-66, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25538066

RESUMO

OBJECTIVES: In present study, we plan to produce a decellularization protocol from rat liver to generate a three-dimensional whole organ scaffold. METHODS: A combination of 1% SDS and 1% tritonX-100 were used orderly to decellularize rat livers. After about 6 h of interactive antegrade/retrograde perfusion, a decellularized whole translucent liver scaffold with integrated blood vessel networks was generated. The decellularized livers are charactered by light microscopy, scanning electron microscopy, and biochemical analysis (DNA quantification) for preservation of the three-dimension of extracellular matrix architecture. RESULTS: The decellularization protocol was verified by observation of the whole translucent liver organ with intact vascular trees under macroscopy, in conjunction with the hematoxylin-eosin staining that showed no cells or nuclear material remained. Additionally, the Masson's stain indicted that the extracellular proteins were well kept and scanning electron microscopy (SEM) revealed a preserved decellularized matrix architecture. Compared to normal livers, DNA in the decellularized livers was quantified less than 10% at the same mass. CONCLUSIONS: The current method of decellularization protocol was feasible, simple and quick, and was verified by an absence of residual cells. The decellularized extracellular matrix had preserved integrate vascular network and a three-dimensional architecture.


Assuntos
Matriz Extracelular/química , Fígado Artificial , Fígado/química , Fígado/ultraestrutura , Engenharia Tecidual/instrumentação , Alicerces Teciduais , Animais , Sistema Livre de Células , Desenho de Equipamento , Análise de Falha de Equipamento , Matriz Extracelular/ultraestrutura , Feminino , Ratos , Ratos Sprague-Dawley
10.
Cell Prolif ; 48(2): 157-65, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25656979

RESUMO

End-stage liver disease can be the termination of acute or chronic liver diseases, with manifestations of liver failure; transplantation is currently an effective treatment for these. However, transplantation is severely limited due to the serious lack of donors, expense, graft rejection and requirement of long-term immunosuppression. Mesenchymal stem cells (MSCs) have attracted considerable attention as therapeutic tools as they can be obtained with relative ease and expanded in culture, along with features of self-renewal and multidirectional differentiation. Many scientific groups have sought to use MSCs differentiating into functional hepatocytes to be used in cell transplantation with liver tissue engineering to repair diseased organs. In most of the literature, hepatocyte differentiation refers to use of various additional growth factors and cytokines, such as hepatocyte growth factor (HGF), fibroblast growth factor (FGF), epidermal growth factor (EGF), oncostatin M (OSM) and more, and most are involved in signalling pathway regulation and cell-cell/cell-matrix interactions. Signalling pathways have been shown to play critical roles in embryonic development, tumourigenesis, tumour progression, apoptosis and cell-fate determination. However, mechanisms of MSCs differentiating into hepatocytes, particularly signalling pathways involved, have not as yet been completely illustrated. In this review, we have focused on progress of signalling pathways associated with mesenchymal stem cells differentiating into hepatocytes along with the stepwise differentiation procedure.


Assuntos
Diferenciação Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos , Hepatócitos/citologia , Células-Tronco Mesenquimais/citologia , Proliferação de Células , Doença Hepática Terminal/terapia , Humanos , Fígado/metabolismo , Transplante de Células-Tronco Mesenquimais , Transdução de Sinais , Engenharia Tecidual
11.
Biomed Mater Eng ; 22(1-3): 105-11, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22766708

RESUMO

Liver diseases have become one of the most important causes of morbidity and mortality in the world. Cell therapy and liver transplantation are though to be two treatment options well accepted. However, the shortage of cells sources in cytotherapy and the lack of liver donor in liver transplantation are the major obstacles for the performance of these treatment methods. It urged us to find new origins of extra-hepatic cells. A number of recent studies show that extra-hepatic mesenchymal stem cells (MSC) from different tissues can be differentiated into hepatocytes like cells (HLC). Several hepatic differentiation protocols of MSC have been published in recent years, based on cellular stimulation with exogenous cytokines/growth factors, co-culture with fetal or adult hepatocytes, 2- or 3-dimensional (2D, 3D) matrices to favor differentiation. Independently from the starting stem cells population used, some minimal criteria must be fulfilled to ensure therapeutic success: in vitro expandability, expression of hepatic like surface markers, with hepatic cell functions, and minimal or absent immunogenicity in the recipient host. In this review, we focused on stem cells originated from bone marrow, umbilical cord and adipose tissue which are widely investigated in recent years and have been proved to have liver regenerative potential, the factors used to differentiate stem cells to hepatocyte-like cells and the methods used to investigate these cells.


Assuntos
Hepatócitos/citologia , Regeneração Hepática , Fígado/fisiologia , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/citologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Humanos , Fígado/citologia , Cordão Umbilical/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA