Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Diabetes Res ; 2015: 257230, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25977928

RESUMO

BACKGROUND: Although numerous reports addressing pathological involvements of diabetic polyneuropathy have been conducted, a universally effective treatment of diabetic polyneuropathy has not yet been established. Recently, regenerative medicine studies in diabetic polyneuropathy using somatic stem/progenitor cell have been reported. However, the effectiveness of these cell transplantations was restricted because of their functional and numerical impairment in diabetic objects. Here, we investigated the efficacy of treatment for diabetic polyneuropathy using angioblast-like cells derived from mouse embryonic stem cells. METHODS AND RESULTS: Angioblast-like cells were obtained from mouse embryonic stem cells and transplantation of these cells improved several physiological impairments in diabetic polyneuropathy: hypoalgesia, delayed nerve conduction velocities, and reduced blood flow in sciatic nerve and plantar skin. Furthermore, pathologically, the capillary number to muscle fiber ratios were increased in skeletal muscles of transplanted hindlimbs, and intraepidermal nerve fiber densities were ameliorated in transplanted plantar skin. Transplanted cells maintained their viabilities and differentiated to endothelial cells and smooth muscle cells around the injection sites. Moreover, several transplanted cells constructed chimeric blood vessels with recipient cells. CONCLUSIONS: These results suggest that transplantation of angioblast like cells induced from embryonic stem cells appears to be a novel therapeutic strategy for diabetic polyneuropathy.


Assuntos
Vasos Sanguíneos/citologia , Diferenciação Celular , Neuropatias Diabéticas/terapia , Transplante de Células-Tronco/métodos , Animais , Diabetes Mellitus Experimental/fisiopatologia , Neuropatias Diabéticas/fisiopatologia , Células-Tronco Embrionárias , Masculino , Camundongos , Condução Nervosa/fisiologia , Nervo Isquiático/fisiopatologia
2.
Biomed Res Int ; 2013: 259187, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24319678

RESUMO

BACKGROUND: Although pathological involvements of diabetic polyneuropathy (DPN) have been reported, no dependable treatment of DPN has been achieved. Recent studies have shown that mesenchymal stem cells (MSCs) ameliorate DPN. Here we demonstrate a differentiation of induced pluripotent stem cells (iPSCs) into MSC-like cells and investigate the therapeutic potential of the MSC-like cell transplantation on DPN. RESEARCH DESIGN AND METHODS: For induction into MSC-like cells, GFP-expressing iPSCs were cultured with retinoic acid, followed by adherent culture for 4 months. The MSC-like cells, characterized with flow cytometry and RT-PCR analyses, were transplanted into muscles of streptozotocin-diabetic mice. Three weeks after the transplantation, neurophysiological functions were evaluated. RESULTS: The MSC-like cells expressed MSC markers and angiogenic/neurotrophic factors. The transplanted cells resided in hindlimb muscles and peripheral nerves, and some transplanted cells expressed S100 ß in the nerves. Impairments of current perception thresholds, nerve conduction velocities, and plantar skin blood flow in the diabetic mice were ameliorated in limbs with the transplanted cells. The capillary number-to-muscle fiber ratios were increased in transplanted hindlimbs of diabetic mice. CONCLUSIONS: These results suggest that MSC-like cell transplantation might have therapeutic effects on DPN through secreting angiogenic/neurotrophic factors and differentiation to Schwann cell-like cells.


Assuntos
Neuropatias Diabéticas/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Indutores da Angiogênese , Animais , Biomarcadores/metabolismo , Glicemia/metabolismo , Peso Corporal , Capilares/patologia , Diferenciação Celular , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Neuropatias Diabéticas/patologia , Neuropatias Diabéticas/fisiopatologia , Extremidades/irrigação sanguínea , Extremidades/patologia , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Fibras Musculares Esqueléticas/patologia , Fatores de Crescimento Neural/metabolismo , Condução Nervosa , Percepção , Nervos Periféricos/patologia , Nervos Periféricos/fisiopatologia , Fluxo Sanguíneo Regional , Pele/irrigação sanguínea , beta-Galactosidase/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 33(9): 2218-21, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23868941

RESUMO

OBJECTIVE: Endothelial damage is an early requisite step for atherosclerosis after vascular injury. It has been reported that vascular wall cells can develop from induced pluripotent stem (iPS) cell-derived fetal liver kinase-1-positive (Flk-1(+)) cells. Here, we investigated the efficacies of intravenously administered iPS cell-derived Flk-1(+) cells on reendothelialization and neointimal hyperplasia in a mouse model of vascular injury. APPROACH AND RESULTS: Femoral arteries of KSN nude mice were injured using a steel wire. Mouse iPS cell-derived Flk-1(+) or Flk-1(-) cells were intravenously injected into those mice at 24 hours after vascular injury. Delivery of iPS cell-derived Flk-1(+) cells significantly attenuated neointimal hyperplasia compared with controls. Evans blue staining of the injured vessel revealed that administration of iPS cell-derived Flk-1(+) significantly enhanced reendothelialization compared with the Flk-1(-) cell control group. Recruitment of PKH26-labeled iPS cell-derived Flk-1(+) cells to the site of injury was also detectable. Expression level of CXCR4 in iPS cell-derived Flk-1(+) cells was 7.5-fold higher than that of iPS cell-derived Flk-1(-) cells. Stromal cell-derived factor-1α treatment significantly enhanced adhesion and migration of iPS cell-derived Flk-1(+) cells to the endothelia, but these were not observed in Flk-1(-) cells. CONCLUSIONS: Intravenously administered iPS cell-derived Flk-1(+) cells are recruited to the site of vascular injury, thereby enhancing reendothelialization followed by suppression of neointimal hyperplasia. Administration of iPS cell-derived Flk-1(+) cells is a potentially useful therapeutic means for vascular dysfunction and prevention of restenosis after angioplasty.


Assuntos
Proliferação de Células , Células Endoteliais/transplante , Artéria Femoral/lesões , Células-Tronco Pluripotentes Induzidas/transplante , Lesões do Sistema Vascular/cirurgia , Animais , Adesão Celular , Linhagem Celular , Movimento Celular , Rastreamento de Células/métodos , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Hiperplasia , Células-Tronco Pluripotentes Induzidas/metabolismo , Injeções Intravenosas , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Neointima , Receptores CXCR4/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Lesões do Sistema Vascular/metabolismo , Lesões do Sistema Vascular/patologia , Proteína Vermelha Fluorescente
4.
Sci Rep ; 3: 1418, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23475393

RESUMO

Angiogenic cell therapy represents a novel strategy for ischemic diseases, but some patients show poor responses. We investigated the therapeutic potential of an induced pluripotent stem (iPS) cell sheet created by a novel magnetite tissue engineering technology (Mag-TE) for reparative angiogenesis. Mouse iPS cell-derived Flk-1(+) cells were incubated with magnetic nanoparticle-containing liposomes (MCLs). MCL-labeled Flk-1(+) cells were mixed with diluted extracellular matrix (ECM) precursor and a magnet was placed on the reverse side. Magnetized Flk-1(+) cells formed multi-layered cell sheets according to magnetic force. Implantation of the Flk-1(+) cell sheet accelerated revascularization of ischemic hindlimbs relative to the contralateral limbs in nude mice as measured by laser Doppler blood flow and capillary density analyses. The Flk-1(+) cell sheet also increased the expressions of VEGF and bFGF in ischemic tissue. iPS cell-derived Flk-1(+) cell sheets created by this novel Mag-TE method represent a promising new modality for therapeutic angiogenesis.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Nanopartículas de Magnetita/química , Engenharia Tecidual , Animais , Técnicas de Cultura de Células , Células Cultivadas , Matriz Extracelular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Membro Posterior/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Isquemia/metabolismo , Isquemia/patologia , Isquemia/terapia , Fluxometria por Laser-Doppler , Lipossomos/química , Camundongos , Camundongos Nus , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Cell Transplant ; 22(10): 1767-83, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23051637

RESUMO

Impaired vascularity and nerve degeneration are the most important pathophysiological abnormalities of diabetic polyneuropathy (DPN). Therefore, regeneration of both the vascular and nervous systems is required for the treatment of DPN. The neural crest (NC) is a transient embryonic structure in vertebrates that differentiates into a vast range of cells, including peripheral neurons, Schwann cells, and vascular smooth muscle cells. In this study, we investigated the ability of transplantation of NC-like (NCL) cells derived from aged mouse induced pluripotent stem (iPS) cells in the treatment of DPN. iPS cells were induced to differentiate into neural cells by stromal cell-derived inducing activity (SDIA) and subsequently supplemented with bone morphogenetic protein 4 to promote differentiation of NC lineage. After the induction, p75 neurotrophin receptor-positive NCL cells were purified using magnetic-activated cell sorting. Sorted NCL cells differentiated to peripheral neurons, glial cells, and smooth muscle cells by additional SDIA. NCL cells were transplanted into hind limb skeletal muscles of 16-week streptozotocin-diabetic mice. Nerve conduction velocity, current perception threshold, intraepidermal nerve fiber density, sensitivity to thermal stimuli, sciatic nerve blood flow, plantar skin blood flow, and capillary number-to-muscle fiber ratio were evaluated. Four weeks after transplantation, the engrafted cells produced growth factors: nerve growth factor, neurotrophin 3, vascular endothelial growth factor, and basic fibroblast growth factor. It was also confirmed that some engrafted cells differentiated into vascular smooth muscle cells or Schwann cell-like cells at each intrinsic site. The transplantation improved the impaired nerve and vascular functions. These results suggest that transplantation of NCL cells derived from iPS cells could have therapeutic effects on DPN through paracrine actions of growth factors and differentiation into Schwann cell-like cells and vascular smooth muscle cells.


Assuntos
Neuropatias Diabéticas/cirurgia , Células-Tronco Pluripotentes Induzidas/citologia , Crista Neural/transplante , Animais , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Células Cultivadas , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Neuropatias Diabéticas/etiologia , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Fibras Nervosas/fisiologia , Fator de Crescimento Neural/metabolismo , Crista Neural/citologia , Crista Neural/metabolismo , Neuritos/fisiologia , Receptor de Fator de Crescimento Neural/metabolismo , Nervo Isquiático/irrigação sanguínea , Nervo Isquiático/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Circulation ; 126(14): 1728-38, 2012 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-22929303

RESUMO

BACKGROUND: Acute coronary syndrome is a leading cause of death in developed countries. Follistatin-like 1 (FSTL1) is a myocyte-derived secreted protein that is upregulated in the heart in response to ischemic insult. Here, we investigated the therapeutic impact of FSTL1 on acute cardiac injury in small and large preclinical animal models of ischemia/reperfusion and dissected its molecular mechanism. METHODS AND RESULTS: Administration of human FSTL1 protein significantly attenuated myocardial infarct size in a mouse or pig model of ischemia/reperfusion, which was associated with a reduction of apoptosis and inflammatory responses in the ischemic heart. Administration of FSTL1 enhanced the phosphorylation of AMP-activated protein kinase in the ischemia/reperfusion-injured heart. In cultured cardiac myocytes, FSTL1 suppressed apoptosis in response to hypoxia/reoxygenation and lipopolysaccharide-stimulated expression of proinflammatory genes through its ability to activate AMP-activated protein kinase. Ischemia/reperfusion led to enhancement of bone morphogenetic protein-4 expression and Smad1/5/8 phosphorylation in the heart, and FSTL1 suppressed the increased phosphorylation of Smad1/5/8 in ischemic myocardium. Treating cardiac myocytes with FSTL1 abolished the bone morphogenetic protein-4-stimulated increase in apoptosis, Smad1/5/8 phosphorylation, and proinflammatory gene expression. In cultured macrophages, FSTL1 diminished lipopolysaccharide-stimulated expression of proinflammatory genes via activation of AMP-activated protein kinase and abolished bone morphogenetic protein-4-dependent induction of proinflammatory mediators. CONCLUSIONS: Our data indicate that FSTL1 can prevent myocardial ischemia/reperfusion injury by inhibiting apoptosis and inflammatory response through modulation of AMP-activated protein kinase- and bone morphogenetic protein-4-dependent mechanisms, suggesting that FSTL1 could represent a novel therapeutic target for post-myocardial infarction, acute coronary syndrome.


Assuntos
Modelos Animais de Doenças , Proteínas Relacionadas à Folistatina/administração & dosagem , Isquemia Miocárdica/tratamento farmacológico , Animais , Apoptose/fisiologia , Células Cultivadas , Proteínas Relacionadas à Folistatina/biossíntese , Proteínas Relacionadas à Folistatina/fisiologia , Humanos , Infusões Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Suínos , Resultado do Tratamento
7.
PLoS One ; 7(6): e39562, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761825

RESUMO

Advanced age is associated with decreased stem cell activity. However, the effect of aging on the differentiation capacity of induced pluripotent stem (iPS) cells into cardiovascular cells has not been fully clarified. We investigated whether iPS cells derived from young and old mice are equally capable of differentiating into vascular progenitor cells, and whether these cells regulate vascular responses in vivo. iPS cells from mouse embryonic fibroblasts (young) or 21 month-old mouse bone marrow (old) were used. Fetal liver kinase-1 positive (Flk-1(+)) cells, as a vascular progenitor marker, were induced after 3 to 4 days of culture from iPS cells derived from young and old mice. These Flk-1(+) cells were sorted and shown to differentiate into VE-cadherin(+) endothelial cells and α-SMA(+) smooth muscle cells. Tube-like formation was also successfully induced in both young and old murine Flk-1(+) cells. Next, hindlimb ischemia was surgically induced, and purified Flk-1(+) cells were directly injected into ischemic hindlimbs of nude mice. Revascularization of the ischemic hindlimb was significantly accelerated in mice transplanted with Flk-1(+) cells derived from iPS cells from either young or old mice, as compared to control mice as evaluated by laser Doppler blood flowmetry. The degree of revascularization was similar in the two groups of ischemic mice injected with iPS cell-derived Flk-1(+) cells from young or old mice. Transplantation of Flk-1(+) cells from both young and old murine iPS cells also increased the expression of VEGF, HGF and IGF mRNA in ischemic tissue as compared to controls. iPS cell-derived Flk-1(+) cells differentiated into vascular progenitor cells, and regulated angiogenic vascular responses both in vitro and in vivo. These properties of iPS cells derived from old mice are essentially the same as those of iPS cells from young mice, suggesting the functionality of generated iPS cells themselves to be unaffected by aging.


Assuntos
Diferenciação Celular/fisiologia , Células Endoteliais/citologia , Endotélio Vascular/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Neovascularização Fisiológica/fisiologia , Fatores Etários , Animais , Linhagem Celular , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Membro Posterior/irrigação sanguínea , Membro Posterior/metabolismo , Isquemia/metabolismo , Masculino , Camundongos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Am J Hypertens ; 25(4): 401-6, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22223040

RESUMO

BACKGROUND: Obesity is a risk factor for the development of cardiovascular diseases that are associated with impaired angiogenesis. Nifedipine, a calcium-channel blocker, has a number of blood pressure (BP)-independent effects as well, such as improving endothelial function and decreasing oxidative stress. Here, we investigated whether nifedipine could improve the angiogenic responses in a diet-induced obese (DIO) model. METHODS: DIO was induced by allowing 8-week-old C57BL/6J mice ad libitum access to a high-fat/high-sucrose (HF/HS) diet. Mice were randomly divided into two groups that were fed either the HF/HS or normal chow. At the age of 12 weeks, the animals were treated/not treated with nifedipine admixed with food at a concentration of 0.001%. Then, 1 week later, the mice were subjected to unilateral hind limb surgery. RESULTS: Angiogenic repair of the ischemic hind limb was impaired in the DIO mice as compared with that in the control mice as evaluated by laser Doppler blood flowmetry (LDBF) and capillary density analysis. Treatment with nifedipine accelerated angiogenic repair in the DIO mice to a level equal to that seen in the control mice. DIO mice showed increased reactive oxygen species (ROS) production after hind limb ischemia. The number of endothelial progenitor cells (EPCs), which contribute to blood vessel formation, was also significantly lower in these mice. Nifedipine treatment ameliorated the oxidative status and increased the number of EPCs in the DIO mice. CONCLUSIONS: Our observations demonstrated that DIO impaired revascularization in response to tissue ischemia. Nifedipine ameliorated obesity-impaired revascularization through suppressing oxidative stress and enhancing the number of EPCs.


Assuntos
Membro Posterior/irrigação sanguínea , Isquemia/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Nifedipino/uso terapêutico , Obesidade/complicações , Animais , Bloqueadores dos Canais de Cálcio/uso terapêutico , Capilares/fisiopatologia , Carboidratos da Dieta/administração & dosagem , Gorduras na Dieta/administração & dosagem , Células Endoteliais/efeitos dos fármacos , Isquemia/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio , Células-Tronco/efeitos dos fármacos
9.
Arterioscler Thromb Vasc Biol ; 31(10): 2210-5, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21757660

RESUMO

OBJECTIVE: Therapeutic angiogenesis with cell transplantation represents a novel strategy for severe ischemic diseases. However, some patients have poor response to such conventional injection-based angiogenic cell therapy. Here, we investigated a therapeutic potential of mesenchymal stem cell (MSC) sheet created by a novel magnetite tissue engineering technology for reparative angiogenesis. METHODS AND RESULTS: Human MSCs incubated with magnetic nanoparticle-containing liposomes were cultured, and a magnet was placed on the reverse side. Magnetized MSCs formed multilayered cell sheets according to magnetic force. Nude mice were subjected to unilateral hind limb ischemia and separated into 3 groups. For the control group, saline was injected into ischemic tissue. In the MSC-injected group, mice received magnetized MSCs by conventional needle injections without sheet formula as a control cell group. In the MSC-sheet group, MSC sheet was layered onto the ischemic tissues before skin closure. Blood flow recovery and the extent of angiogenesis were assessed by a laser Doppler blood flowmetry and histological capillary density, respectively. The MSC-sheet group had a greater angiogenesis in ischemic tissues compared to the control and MSC-injected groups. The angiogenic and tissue-preserving effects of MSC sheets were attributable to an increased expression of vascular endothelial growth factor and reduced apoptosis in ischemic tissues. In cultured MSCs, magnetic labeling itself inhibited apoptosis via a catalase-like antioxidative mechanism. CONCLUSIONS: MSC sheet created by the novel magnetic nanoparticle-based tissue engineering technology would represent a new modality for therapeutic angiogenesis and tissue regeneration.


Assuntos
Óxido Ferroso-Férrico , Isquemia/cirurgia , Magnetismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Apoptose , Células Cultivadas , Modelos Animais de Doenças , Membro Posterior , Humanos , Isquemia/metabolismo , Isquemia/patologia , Isquemia/fisiopatologia , Fluxometria por Laser-Doppler , Lipossomos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Músculo Esquelético/patologia , Fluxo Sanguíneo Regional , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
BMC Cell Biol ; 11: 72, 2010 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-20860813

RESUMO

BACKGROUND: Induced pluripotent stem (iPS) cells are the novel stem cell population induced from somatic cells. It is anticipated that iPS will be used in the expanding field of regenerative medicine. Here, we investigated whether implantation of fetal liver kinase-1 positive (Flk-1+) cells derived from iPS cells could improve angiogenesis in a mouse hind limb model of ischemia. RESULTS: Flk-1+ cells were induced from iPS cells after four to five days of culture. Hind limb ischemia was surgically induced and sorted Flk-1+ cells were directly injected into ischemic hind limbs of athymic nude mice. Revascularization of the ischemic hind limb was accelerated in mice that were transplanted with Flk-1+ cells compared with control mice, which were transplanted with vehicle, as evaluated by laser Doppler blood flowmetry. Transplantation of Flk-1+ cells also increased expression of VEGF mRNA in ischemic tissue compared to controls. CONCLUSIONS: Direct local implantation of iPS cell-derived Flk-1+ cells would salvage tissues from ischemia. These data indicate that iPS cells could be valuable in the therapeutic induction of angiogenesis.


Assuntos
Extremidades/irrigação sanguínea , Células-Tronco Pluripotentes Induzidas/metabolismo , Isquemia/terapia , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular , Separação Celular , Modelos Animais de Doenças , Extremidades/patologia , Extremidades/cirurgia , Citometria de Fluxo , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/transplante , Isquemia/patologia , Camundongos , Camundongos Nus , Transplante de Células-Tronco , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese
11.
Circ Cardiovasc Interv ; 3(2): 166-73, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20332381

RESUMO

BACKGROUND: Adiponectin plays a protective role in the development of obesity-linked disorders. We demonstrated that adiponectin exerts beneficial actions on acute ischemic injury in mice hearts. However, the effects of adiponectin treatment in large animals and its feasibility in clinical practice have not been investigated. This study investigated the effects of intracoronary administration of adiponectin on myocardial ischemia-reperfusion (I/R) injury in pigs. METHODS AND RESULTS: The left anterior descending coronary artery was occluded in pigs for 45 minutes and then reperfused for 24 hours. Recombinant adiponectin protein was given as a bolus intracoronary injection during ischemia. Cardiac functional parameters were measured by a manometer-tipped catheter. Apoptosis was evaluated by terminal deoxynucleotidyltransferase-mediated dUTP nick end-labeling staining. Tumor necrosis factor-alpha and interleukin-10 transcripts were analyzed by real-time polymerase chain reaction. Serum levels of derivatives of reactive oxygen metabolites and biological antioxidant potential were measured. Adiponectin protein was determined by immunohistochemical and Western blot analyses. Intracoronary administration of adiponectin protein led to a reduction in myocardial infarct size and improvement of left ventricular function in pigs after I/R. Injected adiponectin protein accumulated in the I/R-injured heart. Adiponectin treatment resulted in decreased tumor necrosis factor-alpha and increased interleukin-10 mRNA levels in the myocardium after I/R. Adiponectin-treated pigs had reduced apoptotic activity in the I/R-injured heart and showed increased biological antioxidant potential levels and decreased derivatives of reactive oxygen metabolite levels in the blood stream after I/R. CONCLUSIONS: These data suggest that adiponectin protects against I/R injury in a preclinical pig model through its ability to suppress inflammation, apoptosis, and oxidative stress. Administration of intracoronary adiponectin could be a useful adjunctive therapy for acute myocardial infarction.


Assuntos
Adiponectina/administração & dosagem , Estenose Coronária/tratamento farmacológico , Interleucina-10/biossíntese , Miocárdio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Fator de Necrose Tumoral alfa/biossíntese , Animais , Apoptose/efeitos dos fármacos , Quimioterapia Adjuvante , Estenose Coronária/sangue , Testes de Função Cardíaca , Humanos , Interleucina-10/genética , Modelos Animais , Espécies Reativas de Oxigênio/sangue , Proteínas Recombinantes/administração & dosagem , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Suínos , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA