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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 317(4): H765-H776, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31418583

RESUMO

Induced vascular progenitor cells (iVPCs) were created as an ideal cell type for regenerative medicine and have been reported to positively promote collateral blood flow and improve cardiac function in a rat model of myocardial ischemia. Exosomes have emerged as a novel biomedicine that mimics the function of the donor cells. We investigated the angiogenic activity of exosomes from iPVCs (iVPC-Exo) as a cell-free therapeutic approach for ischemia. Exosomes from iVPCs and rat aortic endothelial cells (RAECs) were isolated using a combination of ultrafiltration and size-exclusion chromatography. Nanoparticle tracking analysis revealed that exosome isolates fell within the exosomal diameter (<150 nm). These exosomes contained known markers Alix and TSG101, and their morphology was validated using transmission electron microscopy. When compared with RAECs, iVPCs significantly increased the secretion of exosomes. Cardiac microvascular endothelial cells and aortic ring explants were pretreated with RAEC-Exo or iVPC-Exo, and basal medium was used as a control. iVPC-Exo exerted an in vitro angiogenic effect on the proliferation, tube formation, and migration of endothelial cells and stimulated microvessel sprouting in an ex vivo aortic ring assay. Additionally, iVPC-Exo increased blood perfusion in a hindlimb ischemia model. Proangiogenic proteins (pentraxin-3 and insulin-like growth factor-binding protein-3) and microRNAs (-143-3p, -291b, and -20b-5p) were found to be enriched in iVPC-Exo, which may mediate iVPC-Exo induced vascular growth. Our findings demonstrate that treatment with iVPC-Exo promotes angiogenesis in vitro, ex vivo, and in vivo. Collectively, these findings indicate a novel cell-free approach for therapeutic angiogenesis.NEW & NOTEWORTHY The results of this work demonstrate exosomes as a novel physiological mechanism by which induced vascular progenitor cells exert their angiogenic effect. Moreover, angiogenic cargo of proteins and microRNAs may define the biological contributors in activating endothelial cells to form a new capillary plexus for ischemic vascular diseases.


Assuntos
Células Progenitoras Endoteliais/transplante , Exossomos/transplante , Células-Tronco Pluripotentes Induzidas/transplante , Isquemia/cirurgia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Proteínas Angiogênicas/metabolismo , Animais , Linhagem Celular , Movimento Celular , Proliferação de Células , Citocinas/metabolismo , Modelos Animais de Doenças , Células Progenitoras Endoteliais/metabolismo , Exossomos/metabolismo , Membro Posterior , Células-Tronco Pluripotentes Induzidas/metabolismo , Isquemia/metabolismo , Isquemia/fisiopatologia , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Esquelético/metabolismo , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Transdução de Sinais
2.
Stem Cell Res Ther ; 11(1): 162, 2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-32321589

RESUMO

BACKGROUND: M2 macrophages and exosomes from adipose-derived stem cells (ASCs) are both reported to promote angiogenesis. However, the possible synergistic effects between exogenous exosomes and endogenous M2 macrophages are poorly understood. METHODS: Exosomes were isolated from conditioned medium of normoxic and hypoxic ASCs using the combined techniques of ultrafiltration and size-exclusion chromatography and were identified with nanoparticle tracking analysis and immunoblotting for exosomal markers. Macrophages were collected from the mouse peritoneal cavity. M1 and M2 macrophages were detected by immunoblotting for the intracellular markers inducible nitric oxide synthase (iNOS) and arginase-1 (Arg-1) and by flow cytometry for the surface markers F4/80, CD86, and CD206. Murine models of Matrigel plug and hindlimb ischemia were employed as in vivo angiogenic assays. RESULTS: When M1 macrophages were treated with exosomes from normoxic ASCs (Nor/Exo), and particularly from hypoxic ASCs (Hyp/Exo), the expression of the M1 marker iNOS decreased, and the M2 marker Arg-1 increased in a time- and dose-dependent manner. Additionally, a decrease in the M1 surface marker CD86 and an increase in the M2 surface marker CD206 were observed, which suggested that M1 macrophages were polarized to an M2-like phenotype. Conditioned medium from these M2-like macrophages presented lower levels of proinflammatory cytokines and higher levels of proangiogenic factors and promoted endothelial cell proliferation, migration, and tube formation. Furthermore, M2 polarization and angiogenesis were induced upon the administration of exosomes in mouse Matrigel plug and hindlimb ischemia (HLI) models. Interestingly, these exosomal effects were attenuated by using a colony stimulating factor 1 receptor (CSF-1R) inhibitor, BLZ945, in vitro and in vivo. Downregulation of microRNA-21 (miR-21) in hypoxic ASCs reduced the exosomal effects on M2 polarization, Akt phosphorylation, and CSF-1 secretion. A similar reduction in exosomal activity was also observed when exosomes were administered along with BLZ945. CONCLUSION: Our findings provide evidence that exosomes from ASCs polarize macrophages toward an M2-like phenotype, which further enhances the exosomal proangiogenic effects. Exosomal delivery of miR-21 and positive feedback of secreted CSF-1 may be involved in macrophage polarization.


Assuntos
Exossomos , MicroRNAs , Animais , Membro Posterior , Isquemia/terapia , Macrófagos , Camundongos , Células-Tronco
3.
J Mol Med (Berl) ; 97(2): 141-151, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30554258

RESUMO

Ischemic heart disease (IHD) is caused by the narrowing of arteries that work to provide blood, nutrients, and oxygen to the myocardial tissue. The worldwide epidemic of IHD urgently requires innovative treatments despite the significant advances in medical, interventional, and surgical therapies for this disease. Angiogenesis is a physiological and pathophysiological process that initiates vascular growth from pre-existing blood vessels in response to a lack of oxygen. This process occurs naturally over time and has encouraged researchers and clinicians to investigate the outcomes of accelerating or enhancing this angiogenic response as an alternative IHD therapy. Therapeutic angiogenesis has been shown to revascularize ischemic heart tissue, reduce the progression of tissue infarction, and evade the need for invasive surgical procedures or tissue/organ transplants. Several approaches, including the use of proteins, genes, stem/progenitor cells, and various combinations, have been employed to promote angiogenesis. While clinical trials for these approaches are ongoing, microvesicles and exosomes have recently been investigated as a cell-free approach to stimulate angiogenesis and may circumvent limitations of using viable cells. This review summarizes the approaches to accomplish therapeutic angiogenesis for IHD by highlighting the advances and challenges that addresses the applicability of a potential pro-angiogenic medicine.


Assuntos
Isquemia Miocárdica/terapia , Neovascularização Fisiológica , Indutores da Angiogênese/uso terapêutico , Animais , Exossomos/transplante , Terapia Genética/métodos , Coração/fisiopatologia , Humanos , Isquemia Miocárdica/genética , Isquemia Miocárdica/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Transplante de Células-Tronco/métodos
4.
Artigo em Inglês | MEDLINE | ID: mdl-29348795

RESUMO

Angiogenesis is the process through which new blood vessels are formed, while therapeutic angiogenesis aims to promote and control the angiogenic response. Ischemia results from the lack of blood flow with oxygen and nutrients. Therapeutic angiogenesis is crucial in preserving brain tissue and bodily functions after ischemic stroke. Various approaches have been proposed to promote angiogenesis in ischemic diseases. Traditional protein/gene and subsequent stem/progenitor cell approaches have not shown consistent efficacy for ischemic diseases in clinical trials. Exosomes are microparticles secreted from cells and conduct cell-cell communication including stem cell or cancer cell induced pro-angiogenesis. Utilization of exogenous exosomes for the treatment of ischemic diseases is an emerging approach which may prevent certain disadvantages such as easy degradation and tumor formation happened in other strategies. This review highlights recent reports on the use of exosomes as a therapeutic agent to promote angiogenesis in ischemic stroke.

5.
Stem Cell Res Ther ; 8(1): 125, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28583178

RESUMO

Ischemic diseases, the leading cause of disability and death, are caused by the stenosis or obstruction of arterioles/capillaries that is not compensated for by vessel dilatation or collateral circulation. Angiogenesis is a complex process leading to new blood vessel formation and is triggered by ischemic conditions. Adequate angiogenesis, as a compensatory mechanism in response to ischemia, may increase oxygen and nutrient supplies to tissues and protect their function. Therapeutic angiogenesis has been the most promising therapy for treating ischemic diseases. In recent years, stem cell transplantation has been recognized as a new technique with therapeutic angiogenic effects on ischemic diseases. Adipose-derived stem cells, characterized by their ease of acquisition, high yields, proliferative growth, and low immunogenicity, are an ideal cell source. In this review, the characterization of adipose-derived stem cells and the role of angiogenesis in ischemic attack are summarized. The angiogenic effects of adipose-derived stem cells are discussed from the perspectives of in-vitro, in-vivo, and clinical trial studies for the treatment of ischemic diseases, including ischemic cardiac, cerebral, and peripheral vascular diseases and wound healing. The microvesicles/exosomes released from adipose-derived stem cells are also presented as a novel therapeutic prospect for treating ischemic diseases.


Assuntos
Tecido Adiposo/citologia , Isquemia Encefálica/terapia , Coração/fisiopatologia , Isquemia Miocárdica/terapia , Neovascularização Fisiológica , Doença Arterial Periférica/terapia , Células-Tronco/citologia , Cicatrização , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Ensaios Clínicos como Assunto , Humanos , Ratos , Transplante de Células-Tronco/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA