Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Heart Vessels ; 38(4): 606-613, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36522555

RESUMO

Ischemic preconditioning (IPC) describes a phenomenon wherein brief ischemia of the heart induces a potent cardioprotective mechanism against succeeding ischemic insult. Cyclooxygenase-2 (COX-2), a rate-limiting enzyme in prostanoid biosynthesis, is upregulated in the ischemic heart and contributes to IPC. Prostaglandin E2 (PGE2) protects the heart from ischemia-reperfusion (I/R) injury via its receptor subtype EP4. We sought to clarify the role of the PGE2/EP4 system in the late phase of IPC. Mice were subjected to four IPC treatment cycles, consisting of 5 min of occlusion of the left anterior descending coronary artery (LAD). We found that COX-2 mRNA was significantly upregulated in wild-type hearts at 6 h after IPC treatment. Cardiac PGE2 levels at 24 h after IPC treatment were significantly increased in both wild-type mice and mice lacking EP4 (EP4-/-). At 24 h after IPC treatment, I/R injury was induced by 30 min of LAD occlusion followed by 2 h of reperfusion and the cardiac infarct size was determined. The infarct size was significantly reduced by IPC treatment in wild-type mice; a reduction was not observed in EP4-/- mice. AE1-329, an EP4 agonist, significantly reduced infarct size and significantly ameliorated deterioration of cardiac function in wild-type mice subjected to I/R without IPC treatment. Furthermore, AE1-329 significantly enhanced the I/R-induced activation of Akt, a pro-survival kinase. We demonstrated that the PGE2/EP4 system in the heart plays a critical role in the late phase of IPC, partly by augmenting Akt-mediated signaling. These findings clarify the mechanism of IPC and may contribute to the development of therapeutic strategies for ischemic heart disease.


Assuntos
Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio , Traumatismo por Reperfusão Miocárdica , Camundongos , Animais , Proteínas Proto-Oncogênicas c-akt/uso terapêutico , Ciclo-Oxigenase 2 , Prostaglandinas/uso terapêutico
2.
Am J Physiol Heart Circ Physiol ; 320(6): H2438-H2447, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33961504

RESUMO

Adventitial abnormalities including enhanced vasa vasorum malformation are associated with development and vulnerability of atherosclerotic plaque. However, the mechanisms of vasa vasorum malformation and its role in vascular remodeling have not been fully clarified. We recently reported that ninjurin-1 (Ninj1) is a crucial adhesion molecule for pericytes to form matured neovessels. The purpose is to examine if Ninj1 regulates adventitial angiogenesis and affects the vascular remodeling of injured vessels using pericyte-specific Ninj1 deletion mouse model. Mouse femoral arteries were injured by insertion of coiled wire. Four weeks after vascular injury, fixed arteries were decolorized. Vascular remodeling, including intimal hyperplasia and adventitial microvessel formation were estimated in a three-dimensional view. Vascular fragility, including blood leakiness was estimated by extravasation of fluorescein isothiocyanate (FITC)-lectin or FITC-dextran from microvessels. Ninj1 expression was increased in pericytes in response to vascular injury. NG2-CreER/Ninj1loxp mice were treated with tamoxifen (Tam) to induce deletion of Ninj1 in pericyte (Ninj1 KO). Tam-treated NG2-CreER or Tam-nontreated NG2-CreER/Ninj1loxp mice were used as controls. Intimal hyperplasia was significantly enhanced in Ninj1 KO compared with controls. Vascular leakiness was significantly enhanced in Ninj1 KO. In Ninj1 KO, the number of infiltrated macrophages in adventitia was increased, along with the expression of inflammatory cytokines. In conclusion, deletion of Ninj1 in pericytes induces the immature vasa vasorum formation of injured vasculature and exacerbates adventitial inflammation and intimal hyperplasia. Thus, Ninj1 contributes to the vasa vasorum maturation in response to vascular injury and to reduction of vascular remodeling.NEW & NOTEWORTHY Although abnormalities of adventitial vasa vasorum are associated with vascular remodeling such as atherosclerosis, the mechanisms of vasa vasorum malformation and its role in vascular remodeling have not been fully clarified. The present study provides a line of novel evidence that ninjurin-1 contributes to adventitial microvascular maturation during vascular injury and regulates vascular remodeling.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Artéria Femoral/metabolismo , Neointima/genética , Fatores de Crescimento Neural/genética , Pericitos/metabolismo , Vasa Vasorum/metabolismo , Remodelação Vascular/genética , Túnica Adventícia/metabolismo , Túnica Adventícia/patologia , Animais , Artéria Femoral/lesões , Artéria Femoral/patologia , Técnicas de Inativação de Genes , Hiperplasia/genética , Inflamação/genética , Inflamação/metabolismo , Macrófagos/patologia , Camundongos , Neointima/patologia , Neovascularização Fisiológica/genética , Transcriptoma , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Vasa Vasorum/patologia , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/metabolismo , Lesões do Sistema Vascular/patologia
3.
Biochem Biophys Res Commun ; 519(3): 462-468, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31526566

RESUMO

Ninjurin 1 (Ninj1) is identified as a peripheral nerve injury-induced protein. However, the role of Ninj1 in nerve regeneration is unclear. Schwann cells (SCs) and microvasculature are critical for peripheral nerve regeneration. SCs precursors and microvascular pericytes (PCs), which are nerve/glial antigen 2 (NG2)-positive cells are observed in peripheral nervous system. In this study, we investigated the role of Ninj1 in peripheral nerve regeneration using NG2+cell-specific inducible deletion of Ninj1 mouse model. The number of NG2+cells, which were associated with and without microvessels was increased after sciatic nerve crush injury. There was a significant increase in the expression of Ninj1 and EphA7 in the injured nerve tissue. This increase was mostly observed in NG2+cells. Genetic tracing of NG2+cells was performed using tamoxifen (Tam) treatment on NG2CreERT:R26R-tdTomato mice. The sciatic nerve was injured following the Tam-treatment, then tdTomato-expressing SCs were mostly observed in regenerated SCs at 21 days after nerve injury. Ninj1 gene knockout (Ninj1 KO) in NG2+cells was induced using NG2CreERT:Ninj1loxp mice. Tam-treated-NG2CreERT or Tam-nontreated NG2CreERT:Ninj1loxp mice were used as controls. Following Tam-treatment, the sciatic nerve in each group was injured. Ninj1KO significantly attenuated the expression of the myelin binding protein (MBP) as well as the number of myelinated axons. The expression of MBP in cultured SCs was significantly reduced by SiRNA-mediated Ninj1 knockdown (KD). Ninj1KD also attenuated the differentiation of SCs by isolated EphA7+multipotent PCs. The current data indicate that Ninj1 plays a vital role in peripheral nerve regeneration. This is observed particularly in the myelination process of NG2+cells including SCs precursors and multipotent PCs.


Assuntos
Antígenos/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Fatores de Crescimento Neural/metabolismo , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/fisiopatologia , Proteoglicanas/metabolismo , Células de Schwann/metabolismo , Animais , Antígenos/genética , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Fatores de Crescimento Neural/genética , Pericitos/citologia , Pericitos/metabolismo , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/metabolismo , Proteoglicanas/genética
4.
Arterioscler Thromb Vasc Biol ; 38(10): 2358-2370, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30354207

RESUMO

Objective- Angiogenesis, entire step from endothelial cells (ECs) sprouts to vascular maturation, is a critical response to ischemia. To form functional mature vessels, interactions between ECs and pericytes are essential. Ninj1 (ninjurin1) is an adhesion molecule that contributes to the pathogenesis of neuroinflammation. We recently demonstrated that Ninj1 is expressed in pericytes during angiogenesis. However, the role of Ninj1 in angiogenesis under pathophysiological ischemic conditions has not yet been elucidated. Approach and Results- Ninj1 was detected in microvessels, and its expression was enhanced in ischemic tissues after mouse hindlimb ischemia. Knockdown of Ninj1 was performed by injection of biodegradable microspheres releasing Ninj1-small interfering RNA into muscle tissues. Alternatively, pericyte-specific Ninj1 knockout was induced by tamoxifen treatment of NG2-CreERT/Ninj1-flox mice. Ninj1 knockdown/knockout reduced the formation of blood-circulating functional vessels among total CD31+ microvessels within ischemic tissues and subsequently attenuated color Doppler-assessed blood flow recovery. Ninj1 overexpression enhanced expression of Anpt (angiopoietin) 1, whereas Ninj1 knockdown enhanced the endogenous Anpt1 antagonist, Anpt2 expression in pericytes and inhibited the association of pericytes with ECs and subsequent formation of capillary-like structure, that is, EC tube surrounded with pericytes in 3-dimensional gel culture. Conclusions- Our data demonstrate that Ninj1 is involved in the formation of functional matured vessels through the association between pericytes and ECs, resulting in blood flow recovery from ischemia. These findings further the current our understanding of vascular maturation and may support the development of therapeutics for ischemic diseases.


Assuntos
Moléculas de Adesão Celular Neuronais/deficiência , Células Endoteliais/metabolismo , Deleção de Genes , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Fatores de Crescimento Neural/deficiência , Pericitos/metabolismo , Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/genética , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Membro Posterior , Isquemia/genética , Isquemia/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/genética , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Transdução de Sinais
5.
Circ J ; 79(6): 1363-71, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25766274

RESUMO

BACKGROUND: Capillary pericytes (cPCs), the mural cells of microvessels, play an important role in the formation and maintenance of microvessels; however, little is known about the mechanisms of how cPCs regulate angiogenesis. To identify factors that modulate cPC function, genes whose levels were altered in cPCs during neovessel formation were identified through a microarray screen. METHODS AND RESULTS: Ninjurin1 (nerve injury-induced protein, Ninj1) was selected as a candidate factor for angiogenesis regulation. Ninj1 was expressed in capillary cells including endothelial cells (cECs) and was expressed at a higher level in cPCs. Hypoxia induced the gene expression of Ninj1 in addition of vascular endothelial growth factor (VEGF) in cPCs. When cPCs were co-incubated with a thoracic aorta in a three-dimensional Matrigel system, the length of the EC-tubes sprouting from the aorta was increased. Small interfering RNA-mediated downregulation of Ninj1 in cPCs enhanced these cPCs-mediated angiogenic effects, whereas overexpression of Ninj1 attenuated their effects. The production of angiogenic growth factors, such as VEGF and angiopoietin 1, by cPCs was enhanced by the downregulation of Ninj1, and reduced by the overexpression of Ninj1. CONCLUSIONS: Ninj1 is a novel regulator for the angiogenic effect of PCs. Specifically, Ninj1 negatively regulates the formation of neovessels, that is, the EC-tube, by reducing the trophic effects of cPCs.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Células Endoteliais/citologia , Neovascularização Fisiológica/fisiologia , Fatores de Crescimento Neural/fisiologia , Pericitos/citologia , Animais , Aorta Torácica , Capilares , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Moléculas de Adesão Celular Neuronais/biossíntese , Moléculas de Adesão Celular Neuronais/genética , Técnicas de Cultura de Células , Hipóxia Celular , Linhagem Celular Transformada , Linhagem da Célula , Técnicas de Cocultura , Colágeno , Combinação de Medicamentos , Perfilação da Expressão Gênica , Genes Reporter , Membro Posterior/irrigação sanguínea , Células Endoteliais da Veia Umbilical Humana , Humanos , Técnicas In Vitro , Isquemia/patologia , Laminina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese , Miócitos de Músculo Liso , Fatores de Crescimento Neural/antagonistas & inibidores , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/genética , Técnicas de Cultura de Órgãos , Proteoglicanas , Interferência de RNA , RNA Interferente Pequeno/farmacologia
6.
Clin Exp Hypertens ; 37(5): 411-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25856785

RESUMO

BACKGROUND: Combination antihypertensive therapy with an angiotensin receptor blocker (ARB) and a calcium channel blocker (CCB) or diuretics is common. This subanalysis investigated blood pressure (BP) variability in patients receiving ARB-based combination therapy. METHODS: In a prospective, randomized, open-label trial, hypertensive outpatients (≥65 years) who did not achieve their target BP with ARB monotherapy switched to losartan 50 mg/hydrochlorothiazide 12.5 mg (ARB + D) or ARB plus amlodipine 5 mg (ARB + C) for 12 months. Clinic BP and heart rate (HR), measured every 3 months, visit-to-visit variability and seasonal variation were evaluated. RESULTS: No significant between-group differences in average, maximum, or minimum systolic or diastolic BP, or HR, were found. Visit-to-visit BP variability (systolic) was significantly higher in the ARB + D group than in the ARB + C group. When each group was subdivided into two seasonal groups (summer and winter), no significant between-group differences in BP were found. Multivariate regression analyses showed a tendency toward negative correlation between outdoor temperature and urinary albumin:creatinine ratio and estimated glomerular filtration rate at 12 months in the ARB + D group. CONCLUSION: Combination therapy with an ARB plus a CCB may be preferable to that with an ARB plus diuretics for decreasing BP variability. As for seasonal variability, both treatments can be used safely regardless of season.


Assuntos
Anti-Hipertensivos/administração & dosagem , Pressão Sanguínea/fisiologia , Hipertensão/fisiopatologia , Estações do Ano , Idoso , Anlodipino/administração & dosagem , Pressão Sanguínea/efeitos dos fármacos , Quimioterapia Combinada , Feminino , Humanos , Hidroclorotiazida/administração & dosagem , Hipertensão/tratamento farmacológico , Losartan/administração & dosagem , Masculino , Estudos Prospectivos , Resultado do Tratamento
7.
Lab Invest ; 94(12): 1340-54, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25329003

RESUMO

Adventitial microvessels, vasa vasorum in the vessel walls, have an active role in the vascular remodeling, although its mechanisms are still unclear. It has been reported that microvascular pericytes (PCs) possess mesenchymal plasticity. Therefore, microvessels would serve as a systemic reservoir of stem cells and contribute to the tissues remodeling. However, most aspects of the biology of multipotent PCs (mPCs), in particular of pathological microvessels are still obscure because of the lack of appropriate methods to detect and isolate these cells. In order to examine the characteristics of mPCs, we established immortalized cells residing in adventitial capillary growing at the injured vascular walls. We recently developed in vivo angiogenesis to observe adventitial microvessels using collagen-coated tube (CCT), which also can be used as an adventitial microvessel-rich tissue. By using the CCT, CD146- or NG2-positive cells were isolated from the adventitial microvessels in the injured arteries of mice harboring a temperature-sensitive SV40 T-antigen gene. Several capillary-derived endothelial cells (cECs) and PCs (cPCs) cell lines were established. cECs and cPCs maintain a number of key endothelial and PC features. Co-incubation of cPCs with cECs formed capillary-like structure in Matrigel. Three out of six cPC lines, termed capillary mPCs demonstrated both mesenchymal stem cell- and neuronal stem cell-like phenotypes, differentiating effectively into adipocytes, osteoblasts, as well as schwann cells. mPCs differentiated to ECs and PCs, and formed capillary-like structure on their own. Transplanted DsRed-expressing mPCs were resident in the capillary and muscle fibers and promoted angiogenesis and myogenesis in damaged skeletal muscle. Adventitial mPCs possess transdifferentiation potential with unique phenotypes, including the reconstitution of capillary-like structures. Their phenotype would contribute to the pathological angiogenesis associated with vascular remodeling. These cell lines also provide a reproducible cellular tool for high-throughput studies on angiogenesis, vascular remodeling, and regeneration as well.


Assuntos
Capilares/patologia , Pericitos/fisiologia , Regeneração/fisiologia , Vasa Vasorum/citologia , Remodelação Vascular , Animais , Antígenos , Diferenciação Celular , Separação Celular , Células Endoteliais/fisiologia , Camundongos , Camundongos SCID , Neovascularização Fisiológica , Proteoglicanas , Células-Tronco/fisiologia , Transcriptoma
8.
Biochem Biophys Res Commun ; 443(1): 150-5, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24296254

RESUMO

An immature vasa vasorum in the adventitia of arteries has been implicated in induction of the formation of unstable atherosclerotic plaques. Normalization/maturation of the vasa vasorum may be an attractive therapeutic approach for arteriosclerotic diseases. Nerve growth factor (NGF) is a pleotropic molecule with angiogenic activity in addition to neural growth effects. However, whether NGF affects the formation of microvessels in addition to innervation during pathological angiogenesis is unclear. In the present study, we show a new role for NGF in neovessels around injured arterial walls using a novel in vivo angiogenesis assay. The vasa vasorum around arterial walls was induced to grow using wire-mediated mouse femoral arterial injury. When collagen-coated tube (CCT) was placed beside the injured artery for 7-14 days, microvessels grew two-dimensionally in a thin layer on the CCT (CCT-membrane) in accordance with the development of the vasa vasorum. The perivascular nerve was found at not only arterioles but also capillaries in the CCT-membrane. Biodegradable hydrogels containing VEGF and NGF were applied around the injured artery/CCT. VEGF significantly increased the total length and instability of microvessels within the CCT-membrane. In contrast, NGF induced regeneration of the peripheral nerve around the microvessels and induced the maturation and stabilization of microvessels. In an ex vivo nerve-free angiogenesis assay, although NGF potentially stimulated vascular sprouting from aorta tissues, no effects of NGF on vascular maturation were observed. These data demonstrated that NGF had potent angiogenic effects on the microvessels around the injured artery, and especially induced the maturation/stabilization of microvessels in accordance with the regeneration of perivascular nerves.


Assuntos
Artéria Femoral/efeitos dos fármacos , Artéria Femoral/lesões , Microvasos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Vasa Vasorum/fisiologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Indutores da Angiogênese/farmacologia , Animais , Artéria Femoral/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/inervação , Microvasos/fisiologia , Neovascularização Fisiológica/fisiologia , Vasa Vasorum/inervação
9.
Heart Vessels ; 29(6): 867-72, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24281399

RESUMO

We describe three cases of J-wave syndrome in which ventricular fibrillation (VF) was probably induced by corticosteroid therapy. The patients involved were being treated with prednisolone for concomitant bronchial asthma. One of the three patients had only one episode of VF during her long follow-up period (14 years). Two patients had hypokalemia during their VF episodes. Corticosteroids have been shown to induce various types of arrhythmia and to modify cardiac potassium channels. We discuss the possible association between corticosteroid therapy and VF in J-wave syndrome based on the cases we have encountered.


Assuntos
Antiarrítmicos/administração & dosagem , Reanimação Cardiopulmonar/métodos , Desfibriladores Implantáveis , Glucocorticoides/efeitos adversos , Hipopotassemia , Fibrilação Ventricular , Adulto , Asma/tratamento farmacológico , Eletrocardiografia/métodos , Feminino , Glucocorticoides/administração & dosagem , Humanos , Hipopotassemia/induzido quimicamente , Hipopotassemia/complicações , Hipopotassemia/diagnóstico , Masculino , Síndrome , Resultado do Tratamento , Fibrilação Ventricular/diagnóstico , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/terapia
10.
Am J Physiol Heart Circ Physiol ; 305(8): H1158-67, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23934858

RESUMO

Apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional protein that processes DNA-repair function and controls cellular response to oxidative stress. Endothelial progenitor cells (EPCs) are recruited to oxidative stress-rich injured vascular walls and positively contribute to vascular repair and endothelialization. We hypothesized that APE1 functions for EPCs-mediated inhibition of neointima formation in injured vasculature. EPCs isolated from bone marrow cells of C57BL6 mice (12-16 wk old) were able to survive in the presence of hydrogen peroxide (H2O2; up to 1,000 µM) due to the highly expressed reactive oxygen species (ROS) scavengers. However, adhesion capacity of EPCs was significantly inhibited by H2O2 (100 µM) even though an intracellular ROS was retained at small level. An APE1-selective inhibitor or RNA interference-mediated knockdown of endogenous APE1 in EPCs aggravated the H2O2-mediated inhibition of EPCs-adhesion. In contrast, when APE1 was overexpressed in EPCs using an adenovirus harboring the APE1 gene (APE-EPCs), adhesion was significantly improved during oxidative stress. To examine in vivo effects of APE1 in EPCs, APE-EPCs were transplanted via the tail vein after wire-mediated injury of the mouse femoral artery. The number of adherent EPCs at injured vascular walls and the vascular repair effect of EPCs were enhanced in APE-EPCs compared with control EPCs. Among the cellular functions of EPCs, adhesion is especially sensitive to oxidative stress. APE1 enhances in vivo vascular repair effects of EPCs in part through the maintenance of adhesion properties of EPCs. APE1 may be a novel and useful target gene for effective cellular transplantation therapy.


Assuntos
Adesão Celular/fisiologia , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/fisiologia , Células Endoteliais/fisiologia , Neointima/fisiopatologia , Células-Tronco/fisiologia , Animais , Vasos Sanguíneos/lesões , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Células Endoteliais/transplante , Sequestradores de Radicais Livres , Humanos , Peróxido de Hidrogênio/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxidantes/farmacologia , Espécies Reativas de Oxigênio
11.
Circ J ; 77(4): 1053-62, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23257313

RESUMO

BACKGROUND: Prostacyclin (PGI2) enhances angiogenesis, especially in cooperation with bone marrow (BM)-derived endothelial progenitor cells (EPCs). However, the mechanisms of PGI2 in EPC-mediated angiogenesis in vivo remain unclear. The purpose of this study was to clarify the role of PGI2 in EPC-mediated angiogenesis using BM-specific IP deletion mice. METHODS AND RESULTS: Hind limb ischemia (HLI) was induced in wild-type (WT) mice transplanted with IP-deleted BM (WT/BM(IP(-/-)). Recovery of blood flow (RBF) in WT/BM(IP(-/-)) was impaired for 28 days after HLI, whereas RBF in IP(-/-)/BM(WT) was attenuated for up to 7 days compared with WT/BM(WT). The impaired RBF in WT/BM(IP(-/-)) was completely recovered by intramuscular injection of WT EPCs but not IP(-/-) EPCs. The impaired effects of IP(-/-) EPCs were in accordance with reduced formation of capillary and arterioles in ischemic muscle. An ex vivo aortic ring assay revealed that microvessel formation was enhanced by accumulation/adhesion of EPCs to perivascular sites as pericytes. IP(-/-)EPCs, in which expression of integrins was decreased, had impaired production of angiogenic cytokines, adhesion to neovessels and their angiogenic effects. The small-interfering RNA (siRNA)-mediated knockdown of integrin ß1 in WT EPCs attenuated adhesion to microvessels and their in vivo and in vitro angiogenic effects. CONCLUSIONS: PGI2 may induce persistent angiogenic effects in HLI through adhesion of EPCs to perivascular sites of neovessels via integrins in addition to paracrine effects.


Assuntos
Transplante de Medula Óssea , Células Endoteliais/metabolismo , Epoprostenol/metabolismo , Isquemia/terapia , Microcirculação , Neovascularização Fisiológica , Células-Tronco/metabolismo , Animais , Adesão Celular , Modelos Animais de Doenças , Células Endoteliais/patologia , Epoprostenol/genética , Membro Posterior/irrigação sanguínea , Membro Posterior/metabolismo , Membro Posterior/patologia , Isquemia/genética , Isquemia/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pericitos/metabolismo , Pericitos/patologia , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Células-Tronco/patologia
12.
Am J Physiol Heart Circ Physiol ; 301(6): H2169-80, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21963835

RESUMO

Without heart transplantation, a large number of patients with failing hearts worldwide face poor outcomes. By means of cardiomyocyte regeneration, cardiac regeneration therapy is emerging with great promise as a means for restoring loss of cardiac function. However, the limited success of clinical trials using bone marrow-derived cells and myoblasts with heterogeneous constituents, transplanted at a wide range of cell doses, has led to disagreement on the efficacy of cell therapy. It is therefore essential to reevaluate the evidence for the efficacy of cell-based cardiac regeneration therapy, focusing on targets, materials, and methodologies. Meanwhile, the revolutionary innovation of cardiac regeneration therapy is sorely needed to help the millions of people who suffer heart failure from acquired loss of cardiomyocytes. Cardiac regeneration has been used only in limited species or as a developing process in the rodent heart; now, the possibility of cardiomyocyte turnover in the human heart is being revisited. In the pursuit of this concept, the use of cardiac stem/progenitor stem cells in the cardiac niche must be focused to usher in a second era of cardiac regeneration therapy for the severely injured heart. In addition, tissue engineering and cellular reprogramming will advance the next era of treatment that will enable current cell-based therapy to progress to "real" cardiac regeneration therapy. Although many barriers remain, the prevention of refractory heart failure through cardiac regeneration is now becoming a realistic possibility.


Assuntos
Insuficiência Cardíaca/cirurgia , Miócitos Cardíacos/transplante , Regeneração , Medicina Regenerativa/métodos , Transplante de Células-Tronco , Engenharia Tecidual , Animais , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Miócitos Cardíacos/patologia , Recuperação de Função Fisiológica , Resultado do Tratamento
13.
Arterioscler Thromb Vasc Biol ; 30(3): 464-70, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20007911

RESUMO

OBJECTIVE: Endothelial progenitor cells (EPCs) play an important role in the self-healing of a vascular injury by participating in the reendothelialization that limits vascular remodeling. We evaluated whether prostaglandin I(2) plays a role in the regulation of the function of EPCs to limit vascular remodeling. METHODS AND RESULTS: EPCs (Lin(-)cKit(+)Flk-1(+) cells) were isolated from the bone marrow (BM) of wild-type (WT) mice or mice lacking the prostaglandin I(2) receptor IP (IP(-/-) mice). Reverse transcription-polymerase chain reaction analysis showed that EPCs among BM cells specifically express IP. The cellular properties of EPCs, adhesion, migration, and proliferation on fibronectin were significantly attenuated in IP-deficient EPCs compared with WT EPCs. In contrast, IP agonists facilitated these functions in WT EPCs, but not in IP-deficient EPCs. The specific deletion of IP in BM cells, which was performed by transplanting BM cells of IP(-/-) mice to WT mice, accelerated wire injury-mediated neointimal hyperplasia in the femoral artery. Notably, transfused WT EPCs, but not IP-deficient EPCs, were recruited to the injured vessels, participated in reendothelialization, and efficiently rescued the accelerated vascular remodeling. CONCLUSIONS: These findings clearly indicate that the prostaglandin I(2)-IP system is essential for EPCs to accomplish their function and plays a critical role in the regulation of vascular remodeling.


Assuntos
Endotélio Vascular/metabolismo , Epoprostenol/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica/fisiologia , Animais , Adesão Celular , Movimento Celular , Proliferação de Células , Endotélio Vascular/patologia , Fibronectinas/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patologia , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Knockout , Modelos Animais , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/metabolismo , Transdução de Sinais/fisiologia , Túnica Íntima/lesões , Túnica Íntima/metabolismo , Túnica Íntima/patologia
14.
Sci Rep ; 11(1): 19163, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34580402

RESUMO

Sarcopenia is a pathophysiological malfunction induced by skeletal muscle atrophy. Several studies reported an association between sarcopenia-induced cardiac cachexia and poor prognosis in heart disease. However, due to lack of an established animal models, the underlying mechanism of disturbed cardiac repair accompanied with sarcopenia remains poorly understood. Here, we developed a novel sarcopenia-induced cardiac repair disturbance mouse model induced by tail suspension (TS) after cardiac ischemia and reperfusion (I/R). Importantly, we identified a specific exosomal-microRNA marker, miR-16-5p, in the circulating exosomes of I/R-TS mice. Of note, sarcopenia after I/R disturbed cardiac repair and raised the level of circulating-exosomal-miR-16-5p secreting from both the atrophic limbs and heart of TS mice. Likewise, miR-16-5p mimic plasmid disturbed cardiac repair in I/R mice directly. Additionally, in neonatal rat ventricular myocytes (NRVMs) cultured in vitro under hypoxic conditions in the presence of a miR-16-5p mimic, we observed increased apoptosis through p53 and Caspase3 upregulation, and also clarified that autophagosomes were decreased in NRVMs via SESN1 transcript interference-mediated mTOR activation. In conclusion, we show the pro-apoptotic effect of sarcopenia-derived miR-16-5p, which may be behind the exacerbation of myocardial infarction. Therefore, miR-16-5p can be a novel therapeutic target in the context of cardiac repair disturbances in sarcopenia-cachexia.


Assuntos
Exossomos/genética , MicroRNAs/genética , Infarto do Miocárdio/fisiopatologia , Sarcopenia , Animais , Apoptose , Modelos Animais de Doenças , Elevação dos Membros Posteriores , Masculino , Camundongos Endogâmicos C57BL , Regeneração/genética , Regeneração/fisiologia
15.
Stem Cell Res ; 47: 101914, 2020 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-32738632

RESUMO

Skeletal muscle has a capacity for muscular regeneration mediated by satellite cells (SCs) and non-SCs. Although it is proposed that non-SCs are attractive therapeutic targets for dystrophies, the biological properties of these cells remain unclear. We have recently identified novel multipotent pericytes (PCs), capillary stem cells (CapSCs) derived from the microvasculature. In the present study, we determined if CapSCs contributed to myogenic regeneration using muscular dystrophy mouse model. CapSCs were isolated as EphA7+NG2+PCs from the subcutaneous adipose tissues of GFP-transgenic mice. Co-culture with C2C12 myoblast cells showed that CapSCs effectively enhanced myogenesis as compared to controls including EphA7- PCs and adipose stromal cells (ASCs). CapSCs transplanted in cardiotoxin-injured gastrocnemius muscles were well differentiated into both muscle fibers and microvessels, as compared to controls. At three weeks after cell-transplantation into the limbs of the mdx/utrn-/-mouse, CapSCs increased the number of GFP+myofibers along with dystrophin expression and the area size of myofibers, and also enhanced the muscular mass and its performance, assessed by treadmill test as compared to controls. In conclusion, CapSCs have potent myogenic regeneration capacity and improved the pathological condition in a muscular dystrophy mouse. Thus, CapSCs are an attractive cellular source in regenerative therapy for muscular dystrophy.

16.
Stem Cells Transl Med ; 9(1): 120-130, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31471947

RESUMO

The presence of pericytes (PCs) with multipotency and broad distribution along capillary suggests that microvasculature plays a role not only as a duct for blood fluid transport but also as a stem cell niche that contributes to tissue maintenance and regeneration. The lack of an appropriate marker for multipotent PCs still limits our understanding of their pathophysiological roles. We identified the novel marker EphA7 to detect multipotent PCs using microarray analysis of an immortalized PC library. PCs were isolated from microvessels of mouse subcutaneous adipose tissues, then EphA7+ PCs called capillary stem cells (CapSCs) were separated from EphA7- control PCs (ctPCs) using fluorescence-activated cell sorting system. CapSCs had highly multipotency that enabled them to differentiate into mesenchymal and neuronal lineages compared with ctPCs. CapSCs also differentiated into endothelial cells and PCs to form capillary-like structures by themselves. Transplantation of CapSCs into ischemic tissues significantly improved blood flow recovery in hind limb ischemia mouse model due to vascular formation compared with that of ctPCs and adipose stromal cells. These data demonstrate that EphA7 identifies a subpopulation of multipotent PCs that have high angiogenesis and regenerative potency and are an attractive target for regenerative therapies.


Assuntos
Capilares/metabolismo , Isquemia/imunologia , Células-Tronco Multipotentes/metabolismo , Pericitos/metabolismo , Receptor EphA7/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos
17.
Sci Rep ; 9(1): 7823, 2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-31127150

RESUMO

Apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional protein that controls the cellular response to oxidative stress and possesses DNA-repair functions. It has important roles in the progression and outcomes of various diseases; however, its function and therapeutic prospects with respect to kidney injury are unknown. To study this, we activated APE1 during kidney injury by constructing an expression vector (pCAG-APE1), using an EGFP expression plasmid (pCAG-EGFP) as a control. We performed unilateral ureteral obstruction (UUO) as a model of tubulointerstitial fibrosis on ICR mice before each vector was administrated via retrograde renal vein injection. In this model, pCAG-APE1 injection did not produce any adverse effects and significantly reduced histological end points including fibrosis, inflammation, tubular injury, and oxidative stress, as compared to those parameters after pCAG-EGFP injection. qPCR analysis showed significantly lower expression of Casp3 and inflammation-related genes in pCAG-APE1-injected animals compared to those in pCAG-EGFP-injected UUO kidneys. RNA-Seq analyses showed that the major transcriptional changes in pCAG-APE1-injected UUO kidneys were related to immune system processes, metabolic processes, catalytic activity, and apoptosis, leading to normal kidney repair. Therefore, APE1 suppressed renal fibrosis, not only via antioxidant and DNA-repair functions, but also partly by modulating the immune system through multiple pathways including Il6, Tnf, and chemokine families. Thus, therapeutic APE1 modulation might be beneficial for the treatment of renal diseases.


Assuntos
DNA Liase (Sítios Apurínicos ou Apirimidínicos)/uso terapêutico , Vetores Genéticos/administração & dosagem , Imunidade/genética , Túbulos Renais/patologia , Nefrite Intersticial/terapia , Animais , Biologia Computacional , Reparo do DNA/imunologia , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Modelos Animais de Doenças , Fibrose , Vetores Genéticos/genética , Humanos , Injeções Intravenosas , Túbulos Renais/imunologia , Masculino , Camundongos , Nefrite Intersticial/genética , Nefrite Intersticial/imunologia , Nefrite Intersticial/patologia , Estresse Oxidativo/genética , Estresse Oxidativo/imunologia , Plasmídeos/administração & dosagem , Plasmídeos/genética , RNA-Seq , Veias Renais
19.
Atherosclerosis ; 187(2): 336-42, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16214148

RESUMO

OBJECTIVE: Hyperinsulinemia is a well known risk factor for cardiovascular event. However, it is not known whether hyperinsulinemia facilitates atherosclerotic complex lesions of aorta in non-diabetic patients. We investigated whether hyperinsulinemia is an independent marker of severity of atherosclerosis in thoracic aorta of non-diabetic patients using multiplane transesophageal echocardiography (TEE). RESEARCH DESIGN AND METHODS: Non-diabetic 90 patients with cardiovascular disease underwent TEE, and were analyzed for plasma insulin levels of oral glucose tolerance test, conventional atherosclerotic risk factors and coronary angiographic features. RESULTS: Thoracic aortic plaques were detected in 84 patients (93%). The complex atherosclerotic lesions were observed in 35 (39%) patients, most frequently at the part of aortic arch (p<0.005), showing the greatest atheroma score in thoracic aorta (p<0.05). Univariate analysis showed age, male gender, smoking, coronary artery disease, HDL-cholesterol, insulin levels in glucose tolerance test and homeostasis model assessment insulin resistance index (HOMA index) were found to be significant predictors of complex atherosclerotic lesions. Multivariate regression analysis revealed that HOMA index was an independent predictor of complex atherosclerotic lesions (odds ratio 1.93, p=0.006). There was a significant positive correlation between HOMA index and the atheroma score of thoracic aorta (p<0.001). CONCLUSIONS: Hyperinsulinemia is an independent predictor of complex atherosclerotic lesions detected by TEE in the thoracic aorta of non-diabetic patients.


Assuntos
Aorta Torácica/diagnóstico por imagem , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/epidemiologia , Ecocardiografia Transesofagiana , Hiperinsulinismo/epidemiologia , Idoso , Doenças da Aorta/diagnóstico por imagem , Doenças da Aorta/epidemiologia , Diabetes Mellitus Tipo 2 , Feminino , Humanos , Incidência , Resistência à Insulina , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Valor Preditivo dos Testes , Fatores de Risco , Índice de Gravidade de Doença
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA