Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 35(3): 589-97, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25550202

RESUMO

OBJECTIVE: Arteriogenesis is strongly dependent on the recruitment of leukocytes, especially monocytes, into the perivascular space of growing collateral vessels. On the basis of previous findings that platelets are central players in inflammatory processes and mediate the recruitment of leukocytes, the aim of this study was to assess the role of platelets in a model of arterial remodeling. APPROACH AND RESULTS: C57Bl6 wild-type mice, IL4-R/Iba mice lacking the extracellular domain of the glycoprotein Ibα (GPIbα) receptor, and mice treated with antibodies to block GPIbα or deplete circulating platelets were studied in peripheral arteriogenesis. Using a novel model of intravital 2-photon and epifluorescence imaging, we visualized and quantified the interaction of platelets with leukocytes and the vascular endothelium in vivo. We found that transient platelet adhesion to the endothelium of collateral vessels was a major event during arteriogenesis and depended on GPIbα. Furthermore, leukocyte recruitment was obviously affected in animals with defective platelet GPIbα function. In IL4-R/Iba mice, transient and firm leukocyte adhesion to the endothelium of collateral vessels, as well as leukocyte accumulation in the perivascular space, were significantly reduced. Furthermore, we detected platelet-leukocyte aggregates within the circulation, which were significantly reduced in IL4-R/Iba animals. Finally, platelet depletion and loss of GPIbα function resulted in poor reperfusion recovery as determined by laser Doppler imaging. CONCLUSIONS: Thus, GPIbα-mediated interactions between platelets and endothelial cells, as well as leukocytes, support innate immune cell recruitment and promote arteriogenesis-establishing platelets as critical players in this process.


Assuntos
Neovascularização Fisiológica , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Animais
2.
Am J Physiol Heart Circ Physiol ; 298(3): H754-70, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20023125

RESUMO

The frequently observed de-endothelialization of venous coronary bypass grafts prepared using standard methods exposes subendothelial prothrombotic cells to blood components, thus endangering patients by inducing acute thromboembolic infarction or long-term proliferative stenosis. Our aim was to gain deeper histological and physiological insight into these relations. An intricate network of subendothelial cells, characterized by histological features specific for true pericytes, was detected even in healthy vessels and forms, coupled to the luminal endothelium, a second leaflet of the macrovascular intima. These cells, and particularly those in the venous intima, express enormous concentrations of tissue factor and can recruit additional amounts of up to the 25-fold concentration within 1 h during preincubation with serum (intimal pericytes of venous origin activate 30.71 +/- 4.07 pmol coagulation factor x.min(-1).10(-6) cells; n = 15). Moreover, decoupled from the endothelium, they proliferate rapidly (generation time, 15 +/- 2.1 h, n = 8). Central regions of atherosclerotic plaques, as well as of those of restenosed areas of coronary vein grafts, consist almost completely of these cells. In stark contrast with the prothrombogenicity of the intimal pericytes, intact luminal endothelium recruits high concentrations of thrombomodulin (CD 141) specifically within its intercellular junctions, activates Protein C rapidly (42 +/- 5.1 pmol/min.10(6) venous endothelial cells at thrombin saturation; n = 15), can thus actively prevent coagulatory processes, and never expresses histologically detectable and functionally active tissue factor. Given this strongly prothrombotic potential of the intimal pericytes and their overshooting growth behavior in endothelium-denuded vascular regions, they may play important roles in the development of atherosclerosis, thrombosis, and saphenous vein graft disease.


Assuntos
Pericitos/citologia , Pericitos/fisiologia , Túnica Íntima/citologia , Túnica Íntima/fisiologia , Animais , Bovinos , Técnicas de Cultura de Células/métodos , Proliferação de Células , Separação Celular/métodos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Feminino , Humanos , Modelos Animais , Veia Safena/citologia , Veia Safena/fisiologia , Tromboplastina/metabolismo
3.
FASEB J ; 23(2): 351-61, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18827024

RESUMO

Mobilization of bone marrow-derived stem cells (BMCs) was shown to have protective effects after myocardial infarction (MI). However, the classical mobilizing agent, granulocyte-colony stimulating factor (G-CSF) relapsed after revealing an impaired homing capacity. In the search for superior cytokines, erythropoietin (EPO) appears to be a promising agent. Therefore, we analyzed in a murine model of surgically induced MI the influence of EPO treatment on survival and functional parameters as well as BMC mobilization, homing, and effect on resident cardiac stem cells (CSCs). Human EPO was injected intraperitoneally after ligation of the left anterior descendens (LAD) for 3 days with a total dose of 5000 IU/kg 6 and 30 days after MI, and pressure volume relationships were investigated in vivo. Cardiac tissues were analyzed by histology. To show the effect on BMCs and CSCs, FACS analyses were performed. Homing factors were analyzed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and ELISA. EPO-treated animals showed a significant improvement of survival post-MI (62 vs. 36%). At days 6 and 30, all hemodynamic parameters associated with attenuated remodeling, enhanced neovascularization, and diminished apoptotic cells in the peri-infarct area were improved. BMC subpopulations (CD31(+), c-kit(+), and Sca-1(+) cells) were mobilized, and homing of Sca-1(+) and CXCR4(+) BMCs toward an SDF-1 gradient into the ischemic myocardium was enhanced. However, there was no beneficial effect on CSCs. We have shown that EPO application after MI shows cardioprotective effects. This may be explained by mobilization of BMCs, which are homing via the CXCR-4/SDF-1 axis. However, EPO has no beneficial effects on resident CSCs. Therefore, new treatment regimes using EPO together with other agents may combine complementary beneficial effects preventing ischemic cardiomyopathy.


Assuntos
Células da Medula Óssea/citologia , Cardiomegalia/tratamento farmacológico , Quimiocina CXCL12/metabolismo , Eritropoetina/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Receptores CXCR4/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos CD34/metabolismo , Cardiomegalia/complicações , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Movimento Celular , Eritropoetina/administração & dosagem , Feminino , Imuno-Histoquímica , Antígenos Comuns de Leucócito/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/complicações , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Células-Tronco/citologia , Taxa de Sobrevida , Fatores de Tempo
4.
Antivir Ther ; 14(3): 331-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19474467

RESUMO

BACKGROUND: The aim of this study was to determine the effects of HIV type-1 (HIV-1) infection and antiretroviral therapy (ART) on placental mitochondria. METHODS: HIV-1-infected pregnant women and HIV-1-uninfected controls were enrolled prospectively. Placental mitochondrial DNA (mtDNA) copy numbers were determined by quantitative PCR, subunits II and IV of cytochrome c oxidase (COX) were quantified by western blot and mitochondrial ultrastructure was evaluated by electron microscopy. Venous blood lactate was measured in newborns. RESULTS: In total, 45 HIV-1-infected pregnant women on ART and 32 controls were included. Mean +/-sd mtDNA copy numbers were significantly reduced in ART and HIV-1-exposed placentas (240 +/-118 copies/cell) in comparison with controls (686 +/-842 copies/cell; P<0.001). The mean COX II/IV ratio was 48% lower in the investigational group compared with controls (P<0.001). There was no evidence of severe ultrastructural damage within mitochondria of HIV-1-infected ART-exposed placentas. Although lactate levels between newborns did not differ, they were negatively correlated with placental mtDNA levels. There was no clear association between mitochondrial parameters and a particular nucleoside reverse transcriptase inhibitor (NRTI), the number of NRTIs or time of NRTI exposure. CONCLUSIONS: Placental tissue of HIV-1-infected ART-exposed pregnancies shows evidence of mtDNA depletion with secondary respiratory chain compromise. The clinical effects of this finding warrant further investigation.


Assuntos
Fármacos Anti-HIV/efeitos adversos , Infecções por HIV/tratamento farmacológico , HIV-1 , Mitocôndrias/efeitos dos fármacos , Placenta/efeitos dos fármacos , Complicações Infecciosas na Gravidez/virologia , Adulto , Fármacos Anti-HIV/uso terapêutico , Terapia Antirretroviral de Alta Atividade/efeitos adversos , DNA Mitocondrial/análise , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Humanos , Recém-Nascido/sangue , Ácido Láctico/sangue , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Placenta/metabolismo , Placenta/ultraestrutura , Gravidez , Complicações Infecciosas na Gravidez/tratamento farmacológico , Estudos Retrospectivos
5.
Exp Hematol ; 36(9): 1157-66, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18504066

RESUMO

OBJECTIVE: Cytokine-mediated mobilization of hematopoietic stem cells has become an established method in the field of autologous and allogenic stem cell transplantation. Furthermore, it presents a new concept in tissue repair and regenerative medicine. In the present study, we explored the potency of parathyroid hormone (PTH) compared to granulocyte colony-stimulating factor (G-CSF) for mobilization of stem cells and its regenerative capacity on bone marrow. MATERIALS AND METHODS: Healthy mice were either treated with PTH, G-CSF, or saline. Laboratory parameters were analyzed using a hematological cell analyzer. Hematopoietic stem cells characterized by lin(-)/Sca-1(+)/c-kit(+), as well as subpopulations (CD31(+), c-kit(+), Sca-1(+), CXCR4(+)) of CD45(+)/CD34(+) and CD45(+)/CD34(-) cells were measured by flow cytometry. Immunohistology as well as fluorescein-activated cell sorting analyses were utilized to determine the composition and cell-cycle status of bone marrow cells. Serum levels of distinct cytokines (G-CSF, vascular endothelial growth factor [VEGF]) were determined by enzyme-linked immunosorbent assay. Further, circulating cells were measured after PTH treatment in combination with G-CSF or a G-CSF antibody. RESULTS: Stimulation with PTH showed a significant increase of all characterized subpopulations of bone marrow-derived progenitor cells (BMCs) in peripheral blood (1.5- to 9.8-fold) similar to G-CSF. In contrast to G-CSF, PTH treatment resulted in an enhanced cell proliferation with a constant level of lin(-)/Sca-1(+)/c-kit(+) cells and CD45(+)/CD34(+) subpopulations in bone marrow. Interestingly, PTH application was associated with increased serum levels of G-CSF (2.8-fold), whereas VEGF showed no significant changes. Blocking endogenous G-CSF with an antibody significantly reduced the number of circulating cells after PTH treatment. A combination of PTH and G-CSF showed slight additional effects compared to PTH or G-CSF alone. CONCLUSION: PTH induces mobilization of progenitor cells effectively, which can be related to an endogenous release of G-CSF. In contrast to G-CSF treatment, PTH does not result in a depletion of bone marrow, which may be mediated by an activation of PTH receptor on osteoblasts. The novel function of PTH on mobilization and regeneration of BMCs may pave the way for new therapeutic options in bone marrow and stem cell transplantation as well as in the field of ischemic disorders.


Assuntos
Medula Óssea/efeitos dos fármacos , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Hormônio Paratireóideo/fisiologia , Animais , Células da Medula Óssea/classificação , Ciclo Celular/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/sangue , Fator Estimulador de Colônias de Granulócitos/farmacologia , Imunofenotipagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hormônio Paratireóideo/farmacologia , Fator A de Crescimento do Endotélio Vascular/sangue
6.
Cardiovasc Res ; 77(4): 722-31, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18055578

RESUMO

AIMS: An ongoing concept is that stem cells have the potential to regenerate the injured myocardium. In addition to direct vasorelaxing effects on the vasculature, which are mediated by an increased cAMP production leading to a decreased calcium influx in smooth muscle cells, parathyroid hormone (PTH) was recently shown to facilitate stem cell mobilization. Therefore, we analysed in a murine model of experimental myocardial infarction (MI) the influence of PTH treatment on survival, functional parameters, stem cell migration, and expression of vascular endothelial growth factor A (VEGF-A). METHODS AND RESULTS: Mice (C57BL/6) were treated with PTH (80 microg/kg/d) for up to 14 days after coronary artery ligation. Functional and immunohistochemical analyses were performed at days 6 and 30 after MI. Stem cells and VEGF expression in the myocardium were analysed by FACS and qRT-PCR at day 2 after MI. PTH-treated animals revealed a significant improvement of post-MI survival and myocardial function that was related to a subsequent reduction of left ventricular wall thinning and scar extension. Infarcted hearts of PTH-treated mice revealed increased numbers of CD45(+)/CD34(+) progenitor cells as well as an upregulation of VEGF-A mRNA associated with increased neovascularization and cell survival. CONCLUSIONS: PTH application after MI increases migration of angiogenic CD45(+)/CD34(+) progenitor cells to the ischaemic heart, which may attenuate ischaemic cardiomyopathy. As PTH is already used in patients with osteoporosis, our findings may have a direct impact on the initiation of clinical studies in patients with ischaemic heart disease.


Assuntos
Fármacos Cardiovasculares/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Células-Tronco/efeitos dos fármacos , Animais , Antígenos CD34/análise , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Citometria de Fluxo , Imuno-Histoquímica , Antígenos Comuns de Leucócito/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Comunicação Parácrina , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/imunologia , Células-Tronco/patologia , Fatores de Tempo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
7.
Int J Med Sci ; 5(3): 152-8, 2008 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-18612369

RESUMO

Cardiomyocytes in pulmonary veins (PVs) have been reported in rodents and humans. In humans they were related to atrial arrhythmias, including atrial fibrillation (AF). To investigate histological similarities and differences in PV cardiomyocyte localization and distribution, we performed comparative light and electron microscopic studies on humans, rats and mice, and generated a transgenic mouse strain. Results on mice (C57BL/6 and BALBc) and rats (Wistar) revealed that cardiomyocytes regularly extend from the hilus along venous vessels into the lung tissue surrounding individual intrapulmonary veins of varying diameters (70-250 microm). The cardiomyocytes showed the ultrastructure of a normal working myocardium with intact intercalated discs and tightly packed contractile filaments. In both lung and hilus cardiomyocytes were localized either close to the basal lamina of the endothelium or separated from it by smooth muscle cells and/or collagen fibres. In humans (autopsies, n=20) extrapericardiac cardiomyocytes were only found in 23 out of 78 veins and showed an incomplete sleeve at the lung hilus. In addition, cardiomyocytes occurred significantly more often in right than in left veins, however, never in intrapulmonary veins. We discuss the hypothesis that the variance in distribution of PV cardiomyocytes in humans and rodents might reflect the difference in pathogenesis and development of AF.


Assuntos
Miocárdio/ultraestrutura , Miócitos Cardíacos/ultraestrutura , Veias Pulmonares/metabolismo , Animais , Fibrilação Atrial , Clonagem Molecular , Humanos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Miocárdio/citologia , Miócitos Cardíacos/citologia , Ratos , Ratos Wistar
8.
FASEB J ; 20(7): 956-8, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16571777

RESUMO

Granulocyte-colony stimulating factor (G-CSF) has been shown to improve cardiac function after myocardial infarction (MI) by bone marrow cell mobilization and by protecting cardiomyocytes from apoptotic cell death. However, its role in collateral artery growth (arteriogenesis) has not been elucidated. Here, we investigated the effect of G-CSF on arteriolar growth and cardiac function in a murine MI model. Mice were treated with G-CSF (100 microg/kg/day) directly after MI for 5 consecutive days. G-CSF application resulted in a significant increase of circulating mononuclear cells expressing stem cell markers. Arterioles in the border zone of infarcted myocardium showed an increased expression of ICAM-1 accompanied by an accumulation of bone marrow derived cells and a pronounced proliferation of endothelial and smooth muscle cells. Histology of G-CSF treated mice revealed a lower amount of granulation tissue (67.8 vs. 84.4%) associated with a subsequent reduction in free LV wall thinning and scar extension (23.1 vs. 30.8% of LV). Furthermore, G-CSF treated animals showed a significant improvement of post-MI survival (68.8 vs. 46.2%). Pressure-volume relations revealed a partially restored myocardial function at day 30 (EF: 32.5 vs. 17.2%). Our results demonstrate that G-CSF administration after MI stimulates arteriogenesis and attenuates ischemic cardiomyopathy after MI.


Assuntos
Fator Estimulador de Colônias de Granulócitos/farmacologia , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Vasos Coronários/patologia , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/citologia , Miocárdio/metabolismo , Miocárdio/patologia , Subpopulações de Linfócitos T
10.
Int J Cardiol ; 168(4): 3359-69, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23669105

RESUMO

BACKGROUND: Medical stimulation of endogenous progenitor cell circulation may serve as a new therapeutic tool for treatment of acute myocardial infarction. We analyzed the effects of antidiabetic gliptins plus GCSF (granulocyte colony stimulating factor) on myocardial regeneration after myocardial infarction in a mouse model. METHODS AND RESULTS: After surgical LAD-ligation (left anterior descending artery), Sitagliptin/Vildagliptin was applied yielding sufficient blood levels verified by mass spectrometry and significantly reducing activity of dipeptidyl peptidase (DPP) IV. GCSF or saline was administered intraperitoneally for 6 days. We assessed stem cell mobilization and homing (flow cytometry), infarct size (histology), neovascularization and cellular proliferation (immunohistology), heart function (Millar tip catheterization) and survival (Kaplan-Meier-curves). Gliptins±GCSF administration increased mobilization and cardiac homing of bone-marrow derived stem cells by stabilization of cardiac SDF1 (stromal cell-derived factor). For Sitagliptin, it could be shown that resident cardiac stem cells were stimulated, neovascularization was enhanced and cardiac remodeling was reduced. These effects finally improved myocardial function and increased survival for both gliptins. Although gliptins as a mono therapy lead to remarkable effects in a dose dependent manner and were superior to G-CSF mono-therapy, dual application of GCSF and gliptins revealed the best results. Since both gliptins yielded comparable effects concerning stem cell homing, cardiac function and survival, we suggest a class-effect of DPP-IV-inhibitors. CONCLUSIONS: Thus, gliptins+GCSF and in high concentrations even as mono therapy have beneficial effects on cardiac regeneration after myocardial infarction beyond its anti-diabetic potential.


Assuntos
Adamantano/análogos & derivados , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Coração/efeitos dos fármacos , Hipoglicemiantes/administração & dosagem , Infarto do Miocárdio/tratamento farmacológico , Nitrilas/administração & dosagem , Pirazinas/administração & dosagem , Pirrolidinas/administração & dosagem , Triazóis/administração & dosagem , Adamantano/administração & dosagem , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Quimioterapia Combinada , Coração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfato de Sitagliptina , Vildagliptina
11.
Cardiovasc Res ; 93(2): 330-9, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22080594

RESUMO

AIMS: Parathyroid hormone (PTH) administration after myocardial infarction (MI) is known to attenuate ischaemic cardiomyopathy. This effect mainly resulted from an increase in mobilization and homing of CD34+/CD45+ cells into the ischaemic myocardium. PTH-related stem cell mobilization was shown to be related to endogenous granulocyte-colony stimulating factor (G-CSF) release. The aim of our study is to determine the role of G-CSF on the cardioprotective effects of PTH. METHODS AND RESULTS: G-CSF +/+ (C57BL/6) and G-CSF -/- mice were treated with PTH for 6 days after inducing a MI. The myocardial homing factor stromal cell-derived factor-1 (SDF-1) was analysed on day 2 with enzyme-linked immunosorbent assay. Stem cell populations in peripheral blood and hearts were examined by FACS on days 6 and 2, respectively. Cardiac function and immunohistochemistry were investigated on day 6 and day 30. PTH treatment resulted in a significant increase in CD45+/CD34+ cells in peripheral blood in G-CSF +/+ but not in G-CSF -/- mice. However, a significant increase in SDF-1 and enhanced migration of CD45+/CD34+ cells into the ischaemic myocardium was revealed after PTH administration in both G-CSF +/+ and G-CSF -/- mice. Enhanced stem cell homing was associated with improved cardiac function and post-MI survival after PTH treatment. Furthermore, infarct size, wall thickness, and neovascularization showed a significant improvement in both groups 30 days after MI. CONCLUSION: The cardioprotective effects of PTH were shown to be independent of endogenous G-CSF release and therefore from stem cell mobilization. This puts more emphasis on the role of stem cell homing into ischaemic myocardium.


Assuntos
Fator Estimulador de Colônias de Granulócitos/fisiologia , Coração/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Animais , Apoptose , Células da Medula Óssea/fisiologia , Movimento Celular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isquemia Miocárdica/patologia
12.
Stem Cell Res ; 7(3): 244-55, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21752744

RESUMO

BACKGROUND: G-CSF based stem cell mobilization and stabilization of cardiac SDF-1 by DPP-IV-inhibition (dual stem cell therapy) improve heart function and survival after myocardial infarction. However, it is barely understood whether this new approach acts specifically through the SDF-1/CXCR4 axis, stimulation of resident cardiac stem cells and improved myocardial perfusion. Therefore, we aimed to clarify the role of the SDF1/CXCR4 axis with respect to the benefits of a dual stem cell based therapy. METHODOLOGY/PRINCIPAL FINDINGS: After surgically induced ligation of the LAD, SDF-1/CXCR4 interactions were specifically blocked by the CXCR4 receptor antagonist AMD3100 in G-CSF and Diprotin A treated C57BL/6 mice. G-CSF+DipA treated and non-treated animals served as controls. Because AMD3100 is known to mobilize bone marrow derived stem cells (BMCs) in high concentrations, the optimal dosage (1.25mg per kg body weight) sufficient to block CXCR4 without stimulating mobilization was established. AMD3100 treatment of G-CSF and Diprotin A stimulated mice significantly decreased myocardial homing of circulating stem cells (FACS analysis) and inverted the beneficial effects of (i) cardiac remodeling (histological analyses), (ii) heart function (Millar tip catheterization) and (iii) survival (Kaplan-Meier curves). G-CSF treatment in combination with DPP-IV inhibition enhanced neovascularization at the infarct border zone which was related to an improved myocardial blood flow as measured by SPECT. Moreover, dual stem cell treatment effectively stimulated the pool of resident cardiac stem cells (FACS) which was reversed by AMD3100 treatment. CONCLUSIONS/SIGNIFICANCE: Our data give final proof that homing through the SDF-1/CXCR-4 axis is essential for the success of dual stem cell therapy.


Assuntos
Quimiocina CXCL12/metabolismo , Mobilização de Células-Tronco Hematopoéticas , Infarto do Miocárdio/terapia , Receptores CXCR4/metabolismo , Transplante de Células-Tronco , Animais , Antígenos CD34/metabolismo , Benzilaminas , Quimiocina CXCL12/antagonistas & inibidores , Ciclamos , Dipeptidil Peptidase 4/metabolismo , Relação Dose-Resposta a Droga , Fator Estimulador de Colônias de Granulócitos/farmacologia , Testes de Função Cardíaca/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Modelos Biológicos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Oligopeptídeos/farmacologia , Perfusão , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores CXCR4/antagonistas & inibidores , Análise de Sobrevida
13.
Exp Hematol ; 38(11): 1105-14, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20621157

RESUMO

OBJECTIVE: Clinical studies suggest that transplantation of total bone marrow (BM) after myocardial infarction (MI) is feasible and potentially effective. However, focusing on a defined BM-derived stem cell type may enable a more specific and optimized treatment. Multilineage differentiation potential makes BM-derived multipotent adult progenitor cells (MAPCs) a promising stem cell pool for regenerative purposes. We analyzed the cardioregenerative potential of human MAPCs in a murine model of myocardial infarction. MATERIALS AND METHODS: Human MAPCs were selected by negative depletion of CD45(+)/glycophorin(+) BM cells and plated on fibronectin-coated dishes. In vitro, stem cells were analyzed by reverse transcription polymerase chain reaction. In vivo, we transplanted human MAPCs (5 × 10(5)) by intramyocardial injection after MI in severe combined immunodeficient (SCID) beige mice. Six and 30 days after the surgical procedure, pressure-volume relationships were investigated in vivo. Heart tissues were analyzed immunohistochemically. RESULTS: Reverse transcription polymerase chain reaction experiments on early human MAPC passages evidenced an expression of Oct-4, a stem cell marker indicating pluripotency. In later passages, cardiac markers (Nkx2.5, GATA4, MLC-2v, MLC-2a, ANP, cTnT, cTnI,) and smooth muscle cell markers (SMA, SM22α) were expressed. Transplantation of human MAPCs into the ischemic border zone after MI resulted in an improved cardiac function at day 6 (ejection fraction, 26% vs 20%) and day 30 (ejection fraction, 30% vs 23%). Confirmation of human MAPC marker vimentin in immunohistochemistry demonstrated that human MAPC integrated in the peri-infarct region. The proliferation marker Ki67 was absent in immunohistochemistry and teratoma formation was not found, indicating no tumorous potential of transplanted human MAPCs in the tumor-sensitive SCID model. CONCLUSIONS: Transplantation of human MAPCs after MI ameliorates myocardial function, which may be explained by trophic effects of human MAPCs. Lack of evidence of tumorous potential in the tumor-sensitive SCID model indicates that human MAPCs may deliver an effective and safe stem cell pool for potential treatment of ischemic heart disease.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Multipotentes/citologia , Infarto do Miocárdio/cirurgia , Regeneração/fisiologia , Transplante de Células-Tronco/métodos , Actinas/genética , Adulto , Células-Tronco Adultas/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Antígenos HLA/metabolismo , Hemodinâmica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos SCID , Células-Tronco Multipotentes/metabolismo , Músculo Liso/química , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Fator 3 de Transcrição de Octâmero/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Troponina I/genética , Troponina T/genética
14.
Cell Stem Cell ; 4(4): 313-23, 2009 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-19341621

RESUMO

Ischemic cardiomyopathy is one of the main causes of death, which may be prevented by stem cell-based therapies. SDF-1alpha is the major chemokine attracting stem cells to the heart. Since SDF-1alpha is cleaved and inactivated by CD26/dipeptidylpeptidase IV (DPP-IV), we established a therapeutic concept--applicable to ischemic disorders in general--by combining genetic and pharmacologic inhibition of DPP-IV with G-CSF-mediated stem cell mobilization after myocardial infarction in mice. This approach leads to (1) decreased myocardial DPP-IV activity, (2) increased myocardial homing of circulating CXCR-4+ stem cells, (3) reduced cardiac remodeling, and (4) improved heart function and survival. Indeed, CD26 depletion promoted posttranslational stabilization of active SDF-1alpha in heart lysates and preserved the cardiac SDF-1-CXCR4 homing axis. Therefore, we propose pharmacological DPP-IV inhibition and G-CSF-based stem cell mobilization as a therapeutic concept for future stem cell trials after myocardial infarction.


Assuntos
Quimiocina CXCL12/metabolismo , Inibidores da Dipeptidil Peptidase IV , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Coração/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Infarto do Miocárdio/tratamento farmacológico , Receptores CXCR4/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Coração/fisiologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia
15.
Microvasc Res ; 67(1): 55-63, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14709403

RESUMO

Edema formation is the first manifestation of acute pancreatitis. Microcirculatory derangements like leukocyte-endothelial cell interaction and perfusion failure result in enhancement of microvascular permeability to large molecules playing a pivotal role in the progression of the acutely altered pancreatic tissue. Due to the lack of suitable methods the crucial mechanisms of enhanced permeability in vivo are not very well investigated. Sprague-Dawley rats were randomly assigned to three groups: (a) sham operated animals with normal pancreas, (b) the pancreatitis group induced by 60 min temporary occlusion of the arterial supply followed by reperfusion and (c) the histamine group in which the pancreas was superfused with 10(-5)M histamine. The pharmacokinetics of tetramethylrhodamine-labelled BSA in the intravital microscopic images of a capillary network of the pancreas were densitometrically quantified over 20 min. From these data the effective microvascular permeability was calculated taking also into account morphology of microvessels, elimination rate of the tracer from the intravascular space and capillary microhematocrit. In addition macromolecular leakage of gold-labelled BSA was investigated by electron microscopy. Microvascular permeability was 0.10 +/- 0.02 x 10(-7) cm/s, 0.49 +/- 0.04 x 10(-7) cm/s and 1.21 +/- 0.29 x 10(-7) cm/s for control, ischemia and histamine group, respectively (P < 0.05 ischemia, histamine vs. control and ischemia vs. histamine). Electron microscopy revealed albumin extravasation in the last two groups. We established a technique allowing to quantify microvascular permeability in pancreatic tissue by dynamic intravital microscopy being independent of the investigator. This technique enabling accurate pathophysiologic characterisation in terms of edema formation can form the basis for evaluating in the future novel treatment strategies directed against acute pancreatitis.


Assuntos
Permeabilidade Capilar , Microcirculação/patologia , Microscopia de Fluorescência/métodos , Pâncreas/irrigação sanguínea , Pancreatite/patologia , Animais , Hematócrito , Isquemia , Masculino , Microscopia Eletrônica , Modelos Estatísticos , Perfusão , Permeabilidade , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão , Nitrato de Prata/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA