Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(8)2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38674027

RESUMO

Stem cell therapy stands out as a promising avenue for addressing arthritis treatment. However, its therapeutic efficacy requires further enhancement. In this study, we investigated the anti-arthritogenic potential of human amniotic mesenchymal stem cells (AMM) overexpressing insulin-like growth factor 1 (IGF-1) in a collagen-induced mouse model. The IGF-1 gene was introduced into the genome of AMM through transcription activator-like effector nucleases (TALENs). We assessed the in vitro immunomodulatory properties and in vivo anti-arthritogenic effects of IGF-1-overexpressing AMM (AMM/I). Co-culture of AMM/I with interleukin (IL)-1ß-treated synovial fibroblasts significantly suppressed NF-kB levels. Transplantation of AMM/I into mice with collagen-induced arthritis (CIA) led to significant attenuation of CIA progression. Furthermore, AMM/I administration resulted in the expansion of regulatory T-cell populations and suppression of T-helper-17 cell activation in CIA mice. In addition, AMM/I transplantation led to an increase in proteoglycan expression within cartilage and reduced infiltration by inflammatory cells and also levels of pro-inflammatory factors including cyclooxygenase-2 (COX-2), IL-1ß, NF-kB, and tumor necrosis factor (TNF)-α. In conclusion, our findings suggest that IGF-1 gene-edited human AMM represent a novel alternative therapeutic strategy for the treatment of arthritis.


Assuntos
Artrite Experimental , Edição de Genes , Fator de Crescimento Insulin-Like I , Células-Tronco Mesenquimais , Animais , Humanos , Artrite Experimental/genética , Artrite Experimental/patologia , Artrite Experimental/imunologia , Células-Tronco Mesenquimais/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/genética , Camundongos , Transplante de Células-Tronco Mesenquimais/métodos , Masculino , Camundongos Endogâmicos DBA , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , NF-kappa B/metabolismo , Interleucina-1beta/metabolismo
2.
Int J Mol Sci ; 24(9)2023 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-37175932

RESUMO

Even though the regenerative potential of mesenchymal stem cells (MSCs) has been extensively studied, there is a debate regarding their minimal therapeutic properties. Bone morphogenetic proteins (BMP) are involved in cartilage metabolism, chondrogenesis, and bone healing. In this study, we aimed to analyze the role of genome-edited BMP-2 overexpressing amniotic mesenchymal stem cells (AMMs) in a mouse model of collagen-induced arthritis (CIA). The BMP-2 gene was synthesized and inserted into AMMs using transcription activator-like effector nucleases (TALENs), and BMP-2-overexpressing AMMs (AMM/B) were sorted and characterized using quantitative reverse transcription polymerase chain reaction (qRT-PCR). The co-culture of AMM/B with tumor necrosis factor (TNF)-α-treated synovial fibroblasts significantly decreased the levels of interleukin (IL)-34. The therapeutic properties of AMM/B were evaluated using the CIA mouse model. The injection of AMM/B attenuated CIA progression and inhibited T helper (Th)17 cell activation in CIA mice. In addition, the AMM/B injection increased proteoglycan expression in cartilage and decreased the infiltration of inflammatory cells and factors, including IL-1ß, TNF-α, cyclooxygenase (COX)-2, and Nuclear factor kappa B (NF-kB) in the joint tissues. Therefore, editing the BMP-2 genome in MSCs might be an alternative strategy to enhance their therapeutic potential for treating cartilage degeneration in arthritic joints.


Assuntos
Artrite Experimental , Células-Tronco Mesenquimais , Animais , Humanos , Camundongos , Artrite Experimental/terapia , Artrite Experimental/tratamento farmacológico , Cartilagem/metabolismo , Modelos Animais de Doenças , Fatores Imunológicos/uso terapêutico , Células-Tronco Mesenquimais/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
3.
Int J Mol Sci ; 24(19)2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37834069

RESUMO

Three-dimensional (3D) culture systems have been widely used to promote the viability and metabolic activity of mesenchymal stem cells (MSCs). The aim of this study was to explore the synergistic benefits of using dual 3D MSC culture systems to promote vascular regeneration and enhance therapeutic potential. We used various experimental assays, including dual 3D cultures of human adipose MSCs (hASCs), quantitative reverse transcription polymerase chain reaction (qRT-PCR), in vitro cell migration, Matrigel tube network formation, Matrigel plug assay, therapeutic assays using an ischemic hind limb mouse model, and immunohistochemical analysis. Our qRT-PCR results revealed that fibroblast growth factor 2 (FGF-2), granulocyte chemotactic protein-2 (GCP-2), and vascular endothelial growth factor-A (VEGF-A) were highly upregulated in conventional 3D-cultured hASCs (ASC-3D) than in two-dimensional (2D)-cultured hASCs. Hepatocyte growth factor (HGF), insulin-like growth factor-1 (IGF-1), and stromal-cell-derived factor-1 (SDF-1) showed higher expression levels in cytokine-cocktail-based, 3D-cultured hASCs (ASC-3Dc). A conditioned medium (CM) mixture of dual 3D ASCs (D-3D; ASC-3D + ASC-3Dc) resulted in higher migration and Matrigel tube formation than the CM of single 3D ASCs (S-3D; ASC-3D). Matrigel plugs containing D-3D contained more red blood cells than those containing S-3D. D-3D transplantation into ischemic mouse hind limbs prevented limb loss and augmented blood perfusion when compared to S-3D transplantation. Transplanted D-3D also revealed a high capillary density and angiogenic cytokine levels and transdifferentiated into endothelial-like cells in the hind limb muscle. These findings highlight the benefits of using the dual 3D culture system to optimize stem-cell-based therapeutic strategies, thereby advancing the therapeutic strategy for ischemic vascular disease and tissue regeneration.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Humanos , Camundongos , Animais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Neovascularização Fisiológica/fisiologia , Células-Tronco Mesenquimais/metabolismo , Isquemia/terapia , Isquemia/metabolismo , Meios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Tecido Adiposo/metabolismo
4.
J Cell Mol Med ; 26(13): 3726-3735, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35615995

RESUMO

Although stem cells have extensively been studied as a novel vehicle for tissue repair, their sustained efficacy remains controversial. In this study, we aimed to investigate the angiogenic potency over time of stromal cell-derived factor-1 (SDF-1) gene-edited amniotic mesenchymal stem cells (AMM/S) in a hindlimb ischaemia model. An SDF-1 transgene was inserted into the AMM cell genome via transcription activator-like effector nuclease (TALEN) mediated knock-in, and cell migration, Matrigel tube formation, and in vivo Matrigel plug assays were performed. AMM/S were also transplanted into hindlimb ischaemia model mice. Blood perfusion, therapeutic potential, histology, capillary density and in vivo angiogenic assays were performed. AMM/S exhibited high expression of the SDF-1 gene, and robustly promoted migration, proliferation and microvascular formation. AMM/S transplantation significantly increased blood perfusion and limb loss prevention compared with AMM. AMM/S also significantly inhibited increased capillary density and expression of angiogenic factors in the ischaemic hindlimb. Our study demonstrated that AMM/S provides a significant therapeutic effect in ischaemic hindlimbs by enhancing angiogenesis.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Neovascularização Fisiológica
5.
Int J Mol Sci ; 23(14)2022 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-35887258

RESUMO

Although stem cells are promising tools for the treatment of arthritis, their therapeutic effects remain controversial. In this study, we investigated the therapeutic properties of interleukin (IL)-10-overexpressing human amniotic mesenchymal stem cells (AMMs) generated via gene editing in a collagen-induced mouse model. IL-10 was inserted into the genomic loci of AMMs via transcription activator-like effector nucleases. In vitro immunomodulatory effects of IL-10-overexpressing AMMs (AMM/I) were evaluated and their anti-arthritogenic properties were determined in collagen-induced arthritis (CIA) mice. Transplantation of AMM/I attenuates CIA progression. In addition, the regulatory T cell population was increased, while T helper-17 cell activation was suppressed by AMM/I administration in CIA mice. Consistently, AMM/I injection increased proteoglycan expression, while reducing inflammation and the expression levels of the pro-inflammatory factors, IL-1 ß, IL-6, monocyte chemoattractant protein-1, and tumor necrosis factor- α, in joint tissues. In conclusion, use of IL-10-edited human AMM/I may be a novel therapeutic strategy for the treatment of arthritis.


Assuntos
Artrite Experimental , Transplante de Células-Tronco Mesenquimais , Âmnio , Animais , Artrite Experimental/genética , Artrite Experimental/metabolismo , Artrite Experimental/terapia , Edição de Genes , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Camundongos , Fator de Necrose Tumoral alfa/metabolismo
6.
Int J Mol Sci ; 23(23)2022 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-36499667

RESUMO

Although stem cells have attracted attention as a novel therapeutic solution for tissue regeneration, their minimal efficacy remains controversial. In the present study, we aimed to investigate the enhanced therapeutic property of CXCR4/IL-10 dual angiogenic/anti-inflammatory gene knock-in amniotic mesenchymal stem cells (AMM) in a wound-healing model. Dual CXCR4 and IL-10 genes were inserted into the AMM genome using transcription-activator-like effector nuclease (TALEN). Matrigel tube formation and anti-inflammatory effects were assessed in vitro, and efficacy was tested in vivo in a diabetic wound-healing model. CXCR4/IL-10-expressing amniotic MSCs (AMM/CI) strongly expressed CXCR4 and IL-10 genes and robustly promoted tube formation and anti-inflammatory potential. AMM/CI transplantation resulted in accelerated wound healing, as well as high engraftment and re-epithelialization potential. Transplanted AMM/CI also exhibited high angiogenic and decreased pro-inflammatory gene expression in the wound tissue, indicating direct therapeutic effects on wound healing. Taken together, these data indicate that dual angiogenic/anti-inflammatory gene knock-in may be a novel approach to enhance the therapeutic effects of stem cells, and transplantation of AMM/CI can be an alternative therapeutic option in chronic wound healing.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Humanos , Interleucina-10/metabolismo , Edição de Genes , Âmnio/metabolismo , Cicatrização/genética , Células-Tronco Mesenquimais/metabolismo , Receptores CXCR4/metabolismo
7.
Int J Mol Sci ; 23(1)2021 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-35008610

RESUMO

Previously, we reported the angio-vasculogenic properties of human stromal vascular fraction (SVF) and adipose tissue-derived mesenchymal stem cells (ASCs). In this study, we investigated whether the combination of ASCs and SVF cells exhibited synergistic angiogenic properties. We conducted quantitative (q)RT-PCR, Matrigel plug, tube formation assays, and in vivo therapeutic assays using an ischemic hind limb mouse model. Immunohistochemical analysis was also conducted. qRT-PCR results revealed that FGF-2 was highly upregulated in ASCs compared with SVF, while PDGF-b and VEGF-A were highly upregulated in SVF. Conditioned medium from mixed cultures of ASCs and SVF (A+S) cells showed higher Matrigel tube formation and endothelial cell proliferation in vitro. A+S cell transplantation into ischemic mouse hind limbs strongly prevented limb loss and augmented blood perfusion compared with SVF cell transplantation. Transplanted A+S cells also showed high capillary density, cell proliferation, angiogenic cytokines, and anti-apoptotic potential in vivo compared with transplanted SVF. Our data indicate that A+S cell transplantation results in synergistic angiogenic therapeutic effects. Accordingly, A+S cell injection could be an alternative therapeutic strategy for treating ischemic diseases.


Assuntos
Transplante de Células , Células Endoteliais/fisiologia , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Células-Tronco Mesenquimais , Neovascularização Fisiológica , Fração Vascular Estromal , Animais , Proliferação de Células , Meios de Cultivo Condicionados , Modelos Animais de Doenças , Masculino , Camundongos
8.
J Cardiovasc Pharmacol ; 76(1): 106-111, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32644321

RESUMO

The Cordyceps extract exhibits antiproliferative potential in vascular smooth muscle cells (SMCs) through the mitogen-activated protein kinase signaling pathway. In this study, we aimed to identify the active compounds in the Cordyceps extract and analyze their role in remodeling the arterial wall. On investigation, we discovered the following active compound: 4-methoxyphenyl (E)-3-(furan-3-yl) acrylate and synthesized it. We performed antiproliferation and antimigration assays in addition to an in vivo vessel wall remodeling experiment. Investigation of the mechanism adopted by the active compound to remodel the vessel was performed. The newly synthesized compound inhibited the proliferation and migration of SMCs. Treatment with the synthesized compound reduced neointima formation in the balloon-injured Sprague-Dawley rat model. In addition, this compound inhibited the activation of matrix metalloproteinase-2 and matrix metalloproteinase-9 in type I collagen-activated SMCs. Moreover, this compound suppressed the expression of cycloxygenase-2 (COX-2) in SMCs. Therefore, this compound can exert potential antiarteriosclerotic effects by modulating vessel wall remodeling. In conclusion, the newly synthesized 4-methoxyphenyl (E)-3-(furan-3-yl) acrylate might be an alternative therapeutic intervention for the treatment of atherosclerosis.


Assuntos
Lesões das Artérias Carótidas/prevenção & controle , Proliferação de Células/efeitos dos fármacos , Cordyceps , Furanos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Neointima , Remodelação Vascular/efeitos dos fármacos , Animais , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Cordyceps/química , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Furanos/síntese química , Furanos/isolamento & purificação , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Ratos Sprague-Dawley
9.
J Mol Cell Cardiol ; 126: 118-128, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30500375

RESUMO

Physical microenvironment plays an important role in determining cellular reprogramming. In this study, we first generated directly reprogrammed human dermal fibroblasts (HDFs) into endothelial cells (ECs) mediated by environmental transition-guided cellular reprogramming (e/Entr) using ultrasound and characterized e/Entr. Ultrasound stimulus was introduced to ECs culture media and HDFs and induced into ECs-like cells. We performed microarray, RT-PCR, protein analysis, matrigel plug assay and e/Entr were transplanted into ischemic hindlimb mice model. Here we show that the activation of MAPK signaling pathways and the modulation of histone proteins such as Hp1-α, H3K27me3 and H3K4me3 in e/Entr contribute to the changes in chromatin configuration and reprogramming. Microarray data demonstrated that e/Entr highly expressed genes associated with ECs transcription factors and angiogenesis. In addition, the transplantation of e/Entr into hindlimb ischemia showed a high recovery of blood perfusion, limb salvage and e/Entr contributed to the formation of new vessels. In conclusion, the present study provided the first evidence that ultrasound reprogramming can induce postnatal cells to functional ECs. Therefore, our data suggest that physical stimulus-mediated reprogramming is a highly effective and safe strategy for the novel therapeutic alternatives.


Assuntos
Reprogramação Celular , Fibroblastos/citologia , Células Endoteliais da Veia Umbilical Humana/citologia , Ultrassom , Montagem e Desmontagem da Cromatina , Epigênese Genética , Extremidades/irrigação sanguínea , Humanos , Neovascularização Fisiológica
10.
Cell Physiol Biochem ; 52(4): 935-950, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30964610

RESUMO

BACKGROUND/AIMS: Although mesenchymal stem cells (MSCs) provide effective therapy for liver fibrosis, there are conflicting data regarding their marginal therapeutic effects. This study aimed to enhance the potential of hepatocyte regeneration in human adipose mesenchymal stem cells (ASCs) and investigate whether they have robust therapeutic efficacy in experimental liver fibrosis. METHODS: ASCs were cultured with four cytokines (ASC-C), the expression of hepatogenic factors was detected by microarray, and the effects of conditioned medium (CM) from ASC-C on the activation of hepatic stellate cells were analyzed. The therapeutic effects and mechanism of liver fibrosis induced by thioacetamide (TAA) were determined after cell transplantation. RESULTS: ASC-C exhibited high levels of hepatogenic (HGF, G-CSF), anti-apoptotic (IGFBP-2), and chemokine (IL-8) genes and increased expression of hepatocyte specific proteins. ASC-C CM inhibited the activation of hepatic stellate cells in vitro, and injection of ASC-C significantly delayed TAA-induced liver fibrosis and improved liver function and regeneration in vivo. In addition, human albumin-expressing ASC-C were observed in the livers of recipient animals. High levels of expression of HGF and its downstream signaling molecules, including p-38, were detected in the ASC-C-injected livers. Transplantation of ASC-C exerts anti-fibrotic effects and accelerates liver regeneration. CONCLUSION: Thus, ASC-C may be a novel candidate for the enhanced treatment of liver cirrhosis in clinical settings.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Adipócitos/metabolismo , Fator de Crescimento de Hepatócito/biossíntese , Sistema de Sinalização das MAP Quinases , Transplante de Células-Tronco , Células-Tronco/metabolismo , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/terapia , Adipócitos/patologia , Animais , Linhagem Celular , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco/patologia
11.
Cell Physiol Biochem ; 52(4): 728-741, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30921510

RESUMO

BACKGROUND/AIMS: Stem cells or progenitor cells have been demonstrated as a novel alternative for cell therapy; however, their sustained efficacy is still debated. This study aimed to evaluate whether interleukin 10 (IL-10) gene-edited amniotic mesenchymal stem cells (AMM/I) contribute to left ventricular (LV) function and remodeling after acute myocardial infarction (AMI). METHODS: The IL-10 gene was integrated into the genomic locus of AMM via transcription activator-like effector nucleases (TALEN) and AMM/I were intramyocardially transplanted into AMI mice models. Cardiac function, quantitative polymerase chain reaction, histology, capillary density, and apoptosis assays were performed. RESULTS: AMM/I transplantation significantly suppressed infiltrated CD68 positive or F4/80 positive inflammatory cells and reduced the expression of pro-inflammatory factors in the infarcted myocardium. In addition, significantly improved LV function and reduced infarct size was noted in mice model with AMM/I transplantation than in those given AMM. Moreover, AMM/I highly inhibited cell apoptosis and increased capillary density in the infarcted myocardium. CONCLUSION: Our study demonstrated that AMM/I recruitment played favorable roles in the early restoration of LV function and remodeling by suppressing inflammation and enhancing cardiac protection and capillary density.


Assuntos
Edição de Genes , Interleucina-10/metabolismo , Transplante de Células-Tronco Mesenquimais , Infarto do Miocárdio/terapia , Remodelação Ventricular , Âmnio/citologia , Animais , Análise por Conglomerados , Modelos Animais de Doenças , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Interleucina-10/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/genética , Transcriptoma , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
Acta Cardiol ; 73(1): 13-18, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28885098

RESUMO

OBJECTIVE: The aim of this study is to investigate the molecular mechanisms and effect of rosuvastatin on adhesion molecule induction in human endothelial cells under high-glucose conditions (HG). METHODS AND RESULTS: The effects of rosuvastatin on vascular cell adhesion molecule (VCAM)-1 production and pERK phosphorylation were measured in HG-induced human umbilical vein endothelial cells (HUVECs) with inhibitors targeting the mitogen-activated protein kinase (MAPK) signal pathway. HG increased levels of VCAM-1. Treatment with rosuvastatin inhibited VCAM-1 expression in a concentration- and time-dependent manner. In addition, we investigated the effects of rosuvastatin on the extracellular signal-regulated kinase (ERK) 1/2 signal pathway. Rosuvastatin completely inhibited HG-induced phosphorylation of ERK. ERK/MAPK inhibitors completely prevented the VCAM-1 inhibition effect of rosuvastatin under HG condition. CONCLUSIONS: This study demonstrated that rosuvastatin suppresses HG-induced VCAM-1 production via the MAPK signalling pathway, playing a role in the suppression of atherosclerosis.


Assuntos
Glucose/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Rosuvastatina Cálcica/farmacologia , Regulação para Cima/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/biossíntese , Western Blotting , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Fosforilação
13.
Cytotherapy ; 19(4): 543-554, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28209525

RESUMO

BACKGROUND: Although human stromal vascular fraction (SVF) has been regarded as an attractive stem cell source, its therapeutic mechanism in wound healing has not been fully elucidated. AIMS: In this study, we investigated the molecular characteristics and therapeutic property of SVF for wound healing. METHODS: Microarray data showed that SVF cells are enriched with a higher level of wound healing or epithelium development-related genes and micro RNA. RESULTS: Quantitative polymerase chain reaction (PCR) and reverse transcriptase PCR results revealed that the epithelialization growth factor, epidermal growth factor (EGF), chemokines, stromal cell-derived factor (SDF-1 or CXCL12), neutrophil-activating protein-2 (NAP-2 or CXCL7), chemokine receptors (CXCR1, CCR2 and CCR3) and wound healing genes were up-regulated in SVF compared with those in adipose-derived mesenchymal stem cells (ASCs). An in vitro scratch wound closure experiment demonstrated that co-culture with SVF substantially accelerated the wound closure of fibroblasts. Wounds in nude mice were created by skin excisions followed by injections of SVF with Pluronic hydrogel. SVF implantation highly accelerated wound closure and increased cellularity and re-epithelialization. In addition, the transplanted SVF exhibited high engraftment rates in the wound area, suggesting direct benefits for cutaneous closure. CONCLUSIONS: Taken together, these data suggest that SVF possesses high therapeutic capability for wound healing via the secretion of epithelialization and chemotactic growth factors and enhanced engraftment properties.


Assuntos
Vasos Sanguíneos/citologia , Quimiotaxia , Reepitelização/fisiologia , Células Estromais/fisiologia , Células Estromais/transplante , Animais , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/citologia , Fibroblastos/fisiologia , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Reepitelização/genética , Células Estromais/citologia , Regulação para Cima
14.
Circ J ; 80(4): 998-1007, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26853554

RESUMO

BACKGROUND: Although stem cells have been regarded as a promising therapeutic option, the marginal therapeutic effects of stem cells are limitations that must be overcome for the development of effective cell therapy. This study sought to identify the angio-vasculogenic properties of endothelial differentiated mesenchymal stem cells (MSCs) and to determine whether these cells are effective for vascular repair. METHODS AND RESULTS: Adipose MSCs were cultured for 10 days under endothelial cell (EC) culture conditions. These endothelial cell differentiated adipose MSCs (EA) and undifferentiated adipose MSCs (UA) were characterized via angiogenesis and adhesion assays. These cells were transplanted into a hindlimb ischemia (HLI) model to determine therapeutic effects and their underlying mechanisms. EA displayed low adhesion and angiogenic properties in vitro compared with UA. When implanted into mouse HLI models, EA exhibited the decreased recovery of blood perfusion in limb ischemia than uncultured UA. Histology data showed that injected EA exhibited lower retention, angiogenic cytokine levels, and neovascularization in vivo than did UA. Short-term differentiated EA display less cell engraftment and angio-vasculogenic potential, and are less effective for peripheral vascular repair than UA. CONCLUSIONS: EC differentiation of MSCs may not present an effective strategy for the promotion of therapeutic neovascularization.


Assuntos
Membro Posterior/irrigação sanguínea , Células Endoteliais da Veia Umbilical Humana/metabolismo , Isquemia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica , Animais , Modelos Animais de Doenças , Xenoenxertos , Humanos , Isquemia/metabolismo , Isquemia/patologia , Isquemia/terapia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
15.
Cell Mol Biol Lett ; 21: 24, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28536626

RESUMO

Cordyceps belongs to a genus of acormycete fungi and is known to exhibit various pharmacological effects. The aim of this study was to investigate the effect of Cordyceps species on the proliferation of vascular smooth muscle cells (VSMC) and their underlying molecular mechanism. A cell proliferation assay showed that Cordyceps bassiana ethanol extract (CBEE) significantly inhibited VSMC proliferation. In addition, neointimal formation was significantly reduced by treatment with CBEE in the carotid artery of balloon-injured rats. We also investigated the effects of CBEE on the extracellular signal-regulated kinase (ERK) signal pathway. Western blot analysis revealed increased ERK 1/2 phosphorylation in VSMCs treated with CBEE. Pretreatment with U0126 completely abrogated CBEE-induced ERK 1/2 phosphorylation. In conclusion, CBEE exhibited anti-proliferative properties that affected VSMCs through the ERK1/2 MAPK signaling pathway. Our data may elucidate the inhibitory mechanism of this natural product.


Assuntos
Proliferação de Células/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Extratos de Tecidos/farmacologia , Animais , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Cordyceps/química , Masculino , Miócitos de Músculo Liso/fisiologia , Neointima/tratamento farmacológico , Ratos
16.
J Cell Physiol ; 230(8): 1740-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25215878

RESUMO

Flt3 ligand (FL), a potent hematopoietic cytokine, plays an important role in development and activation of dendritic cells (DCs) and natural killer cells (NK). Although some post-receptor signaling events of FL have been characterized, the role of FL on Flt3 expressing human peripheral blood monocyte is unclear. In the current study, we examined the role of FL on cell survival and growth of peripheral blood monocytes and function of monocyte-derived DCs. FL promoted monocyte proliferation in a dose-dependent manner and prevented spontaneous apoptosis. FL induced ERK phosphorylation and a specific ERK inhibitor completely abrogated FL-mediated cellular growth, while p38 MAPK, JNK, and AKT were relatively unaffected. Addition of FL to GM-CSF and IL-4 during DCs generation from monocytes increased the yield of DCs through induction of cell proliferation. DCs generated in the presence of FL expressed more costimulatory molecules on their surfaces and stimulated allogeneic T cell proliferation in MLR to a higher magnitude. Furthermore, FL partially antagonized IL-10-mediated inhibition on DCs function. Further characterization of FL actions may provide new and important information for immunotherapeutic approaches utilizing DCs.


Assuntos
Células Dendríticas/metabolismo , Proteínas de Membrana/metabolismo , Monócitos/metabolismo , Apoptose/imunologia , Western Blotting , Proliferação de Células , Células Dendríticas/citologia , Células Dendríticas/imunologia , Citometria de Fluxo , Humanos , Imunofenotipagem , Técnicas In Vitro , Ativação Linfocitária/imunologia , Proteínas de Membrana/imunologia , Monócitos/citologia , Monócitos/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Crit Rev Eukaryot Gene Expr ; 25(2): 145-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26080608

RESUMO

Adipose tissue has emerged as an attractive cell source in tissue engineering and regenerative medicine because it can be easily collected and enriched with stem/progenitor cell populations. The stromal vascular fraction (SVF) derived from adipose tissue contains heterogeneous cell populations such as mesenchymal progenitor/stem cells, preadipocytes, endothelial cells, pericytes, T cells, and M2 macrophages. SVF-derived mesenchymal progenitor/stem cells can be easily expanded in vitro and have the potential to create diverse lineages of cells. Although there have been issues related to their isolation and purification, SVF cells demonstrate regenerative potential in damaged tissues or organs through paracrine and differentiation mechanisms. Furthermore, SVF cells augment immunological tolerance by promoting inhibitory macrophages and T regulatory cells and by decreasing ongoing inflammation. Numerous implantations of freshly isolated, autologous adipose tissue-derived SVF cells in cosmetic surgeries and in a wide variety of other specialties support the safety of SVF cells and have accelerated their clinical application. Despite these attractive advantages of SVF cells in clinical interventions, to our knowledge the recent status of clinical studies of various diseases has not been fully investigated. Therefore this article describes recent advances in the clinical use of SVF cells, as well as the associated challenges and future directions for this field of research. We also speculate that verification of the efficacy and safety of SVF cells requires more basic experimental research, using a standard isolation protocol, and larger randomized clinical trials of the utility of SVF cells in various diseases.


Assuntos
Tecido Adiposo/citologia , Células Estromais/metabolismo , Tecido Adiposo/metabolismo , Animais , Diferenciação Celular , Modelos Animais de Doenças , Humanos , Medicina Regenerativa , Células-Tronco , Células Estromais/citologia , Engenharia Tecidual
18.
Cell Mol Biol Lett ; 20(5): 937-47, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26636413

RESUMO

Ramipril has recently been shown to have anti-atherogenic properties. However, the specific mechanisms underlying these effects remain unclear. The purpose of this study was to determine the effects of ramipril on induction of adhesion molecules in human umbilical vein endothelial cells (HUVECs) using high-glucose (HG) conditions and to investigate possible underlying molecular mechanisms. The effects of ramipril on expression of intercellular adhesion molecule (ICAM)-1, vascular cell adhesion molecule (VCAM)-1 production, and ERK phosphorylation were examined in HG-induced HUVECs with inhibitors targeting the mitogen-activated protein kinase (MAPK) signaling pathway. HG induced the expression of the adhesion molecules ICAM-1 and VCAM-1. Pretreatment with ramipril drastically inhibited ICAM-1 and VCAM-1 production in a time- and dose-dependent manner. Moreover, upon investigating the effects of ramipril on the MAPK/extracellular signal-regulated kinase (ERK) signaling pathway, we found that ramipril completely inhibited HG-induced phosphorylation of ERK1/2. ERK inhibitors completely prevented the inhibitory effect of HG. This study demonstrated that ramipril reduces HG-stimulated induction of ICAM-1 and VCAM-1 expression via MAPK signaling, which may be useful for inhibition of atherosclerosis.


Assuntos
Anti-Hipertensivos/farmacologia , Moléculas de Adesão Celular/metabolismo , Glucose/farmacologia , Ramipril/farmacologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia
19.
Mol Cell Neurosci ; 62: 60-7, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25128805

RESUMO

Recently, we reported that human peripheral blood (PB)-derived CD31(+) cells are highly angiogenic. In this study, we investigated the beneficial effects of CD31(+) cells on peripheral neuropathy in mice. CD31(+) cells were collected from the peripheral blood using magnetic activated cell sorting. CD31(+) cells exhibited higher levels of expression of angiogenic genes on real-time reverse transcriptase polymerase chain reaction. Peripheral neuropathy was induced by crushing the sciatic nerve with a hemostat, and CD31(+) cells were then injected intramuscularly along the sciatic nerve. CD31(+) cell transplantation restored motor nerve conduction velocity and voltage amplitude and improved motor coordination. In addition, CD31(+) cell transplantation significantly improved blood perfusion and increased intraneural vascularity in the sciatic nerve. Whole-mount fluorescent imaging and dot blot analysis showed that CD31(+) cells in the nerve possessed high engraftment and anti-apoptotic properties. Additionally, injected CD31(+) cells displayed neurovascular tropism and are highly incorporated with vasculature. Angiogenic cytokines were augmented in CD31(+)-injected nerve tissue, suggesting increased neovascularization. Taken together, these results indicate that CD31(+) cells might be a novel therapeutic strategy in the treatment of peripheral neuropathy.


Assuntos
Transplante de Células , Células Endoteliais/metabolismo , Doenças do Sistema Nervoso Periférico/terapia , Nervo Isquiático/metabolismo , Animais , Transplante de Células/métodos , Terapia Baseada em Transplante de Células e Tecidos , Modelos Animais de Doenças , Células Endoteliais/imunologia , Humanos , Camundongos SCID , Neovascularização Fisiológica/fisiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Nervo Isquiático/lesões
20.
J Cell Mol Med ; 18(11): 2321-30, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25267411

RESUMO

Recently, we reported the properties of CD31-expressing cells in healthy individuals. However, the characteristics of CD31-expressing cells derived from coronary artery disease (CAD) patients remain unknown. This study aimed to investigate the relationship between circulating CD31(+) cells and CAD as well as their biological characteristics. Analysis with flow cytometry revealed that CD31(+) cells (C-CD31) from the peripheral blood (PB) of CAD patients exhibited low levels of T-cell marker and high levels of macrophage marker compared with the PB-CD31(+) cells from healthy individuals (H-CD31). In addition, the expression levels of multiple pro-angiogenic and chemokine genes were significantly down-regulated in C-CD31. However, inflammatory gene IL-1α was highly up-regulated in C-CD31. Patients with unstable angina (UA) had significantly more CD31(+) cells in the PB than healthy control group (P < 0.001). Moreover, there were significant correlations between the number of CD31(+) cells and cardiovascular (CV) disease activity (R = 0.318, P = 0.006) and the number of diseased coronaries (R = 0.312, P = 0.005). For the diagnostic category of UA, the area under curve was 0.803 (P < 0.001). In conclusion, C-CD31 have impaired angiogenic potential and the number of circulating CD31(+) cells were correlated with CV risk. These findings may contribute to the understanding of the pathogenesis of CAD.


Assuntos
Angina Pectoris/sangue , Doença da Artéria Coronariana/sangue , Inflamação/sangue , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Idoso , Angina Pectoris/patologia , Doença da Artéria Coronariana/patologia , Feminino , Citometria de Fluxo , Humanos , Inflamação/patologia , Masculino , Pessoa de Meia-Idade , Molécula-1 de Adesão Celular Endotelial a Plaquetas/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA