Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Biomed Res Int ; 2017: 9634172, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28630875

RESUMO

Glioblastoma is the most aggressive cancer of the brain. The dismal prognosis is largely attributed to the heterogeneous nature of the tumor, which in addition to intrinsic molecular and genetic changes is also influenced by the microenvironmental niche in which the glioma cells reside. The cancer stem cells (CSCs) hypothesis suggests that all cancers arise from CSCs that possess the ability to self-renew and initiate tumor formation. CSCs reside in specialized niches where interaction with the microenvironment regulates their stem cell behavior. The reciprocal interaction between glioma stem cells (GSCs) and cells from the microenvironment, such as endothelial cells, immune cells, and other parenchymal cells, may also promote angiogenesis, invasion, proliferation, and stemness of the GSCs and be likely to have an underappreciated role in their responsiveness to therapy. This crosstalk may also promote molecular transition of GSCs. Hence the inherent plasticity of GSCs can be seen as an adaptive response, changing according to the signaling cue from the niche. Given the association of GSCs with tumor recurrence and treatment sensitivity, understanding this bidirectional crosstalk between GSCs and its niche may provide a framework to identify more effective therapeutic targets and improve treatment outcome.


Assuntos
Glioblastoma/imunologia , Células-Tronco Neoplásicas/imunologia , Neovascularização Patológica/imunologia , Microambiente Tumoral/imunologia , Animais , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia
2.
Eur J Prev Cardiol ; 20(3): 496-512, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22496273

RESUMO

Vascular homeostasis is an integral reflection of quiescent, but competent endothelium. Endothelial microparticles (EMPs) are circulating submicron-sized membranous vesicles released by endothelium that are increasingly recognized and play a multitude of biological roles reflecting competency of endothelial function and vascular biology. EMPs have been shown to act as primary and secondary messengers of vascular inflammation, thrombosis, vasomotor response, angiogenesis, and endothelial survival. These often-neglected vesicles are emerging as potentially useful indicators of dysfunctioning endothelium, whereby differential presence of EMPs is linked to disease manifestation. In this review, we outline the evolving understanding of EMPs from mere passive vesiculation to active messenger in pathophysiological responses. Current evidence implicating EMPs in cardiopulmonary, renal, cerebral, and metabolic disorders warrants vigorous clinical validation. While studies of EMPs face significant technical challenges that await standardization and consolidation, unravelling its biological significance in endothelial dysfunction could herald the advent of novel non-invasive surveillance platforms for vascular health, risk stratification, and disease prognostication.


Assuntos
Doenças Cardiovasculares/metabolismo , Micropartículas Derivadas de Células/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Animais , Biomarcadores/metabolismo , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Micropartículas Derivadas de Células/patologia , Células Endoteliais/patologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Prognóstico , Fatores de Risco , Transdução de Sinais
3.
Cardiovasc Res ; 89(2): 329-35, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20870652

RESUMO

AIMS: In this study, we investigated and compared the electrophysiological and molecular properties of large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels between human internal mammary arteries (IMA) and radial arteries (RA). METHODS AND RESULTS: IMA and RA sections were obtained from 79 patients (including 9 females) undergoing coronary artery bypass graft surgery. We examined the effects of K(+) channel blockers tetraethylammonium (TEA), iberiotoxin (IBTX), and 4-aminopyridine (4-AP) on isolated smooth muscle cells (SMCs) using patch clamping. Both TEA (1 mM) and IBTX (0.1 µM) significantly decreased K(+) currents in IMA SMCs and RA SMCs, while 4-AP (1 mM) only had a weak effect. IBTX had a greater K(+)-blocking effect on IMA SMCs than on RA SMCs. Consistently, TEA and IBTX evoked significant constriction of both intact vascular rings. IBTX had a greater constrictor effect on IMA rings (18.5 ± 6.7%, n= 8) than on RA rings (10.6 ± 3.1%, n= 8), P< 0.05. RT-PCR and western blot analysis demonstrated that gene and protein expression of the α-subunit of BK(Ca) channels from IMA was greater than that from RA. CONCLUSION: The density of BK(Ca) channels is greater in human IMA than in RA resulting in greater BK(Ca) currents in SMCs from IMA. This may partly explain the finding of less spasm in IMA grafts than in RA grafts. Our results may be of value in determining the best anti-spasm agent to use peri-operatively.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Artéria Torácica Interna/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Artéria Radial/metabolismo , Western Blotting , Ponte de Artéria Coronária/efeitos adversos , Feminino , Humanos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Masculino , Artéria Torácica Interna/efeitos dos fármacos , Artéria Torácica Interna/cirurgia , Potenciais da Membrana , Pessoa de Meia-Idade , Músculo Liso Vascular/cirurgia , Miócitos de Músculo Liso/efeitos dos fármacos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Artéria Radial/efeitos dos fármacos , Artéria Radial/cirurgia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espasmo/etiologia , Espasmo/metabolismo , Coleta de Tecidos e Órgãos , Vasoconstrição
4.
J Heart Lung Transplant ; 29(12): 1415-26, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20688539

RESUMO

BACKGROUND: Differentiation of bone marrow stem cells toward cardiomyocytes has been widely reported in vitro. However, optimum cell types and mechanisms leading to functional improvement in cardiac cell therapy remain unresolved. There is limited evidence showing a dose-dependent effect of transplanted cells in contributing to functional recovery. This study showed that cell transplantation of differentiated cardiomyocyte-like cells (CLCs) and undifferentiated mesenchymal stem cells (MSCs) dose-dependently improved left ventricular function in a rat myocardial infarction model. METHODS: At 1 week after infarction in Wistar rats, 1 × 10(6) MSCs (n = 9) or CLCs (n = 9) and 5 × 10(6) MSCs (n = 18) or CLCs (n = 15) were injected into peri-infarcted myocardium to study their effect after 6 weeks. RESULTS: High-dose CLCs exhibited a dose-response that was significantly more effective than MSCs in recovering cardiac contractility. Superiority of CLCs over MSCs was demonstrated in load-independent measurement of the end-systolic pressure-volume relationship and pre-load recruitable stroke work, but not in the end-diastolic pressure-volume relationship. These findings showed a unique systolic role of CLCs in contractility recovery. Functional improvement mediated by MSCs was mainly derived from preservation of endogenous myocyte function and restriction of chamber dilatation by enhancing intramyocardial angiogenesis during post-infarct ventricular remodeling. Engrafted CLCs showed better survival, were strategically integrated into myofiber-associated collagen V matrix, and exhibited mature sarcomeric cross-striations. Vascular differentiation, but not cardiac, was observed with MSCs. CONCLUSION: These cell type-specific effects suggest that committing stem cells to a cardiac phenotype ex vivo promoted mechanical and functional integration of CLCs into the myofibrillar syncytium of infarcted myocardium.


Assuntos
Diferenciação Celular , Transplante de Células-Tronco Mesenquimais , Infarto do Miocárdio/cirurgia , Miócitos Cardíacos/transplante , Função Ventricular Esquerda , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Células-Tronco Mesenquimais , Miócitos Cardíacos/citologia , Ratos , Ratos Wistar , Sístole
5.
Ann Thorac Surg ; 86(6): 1841-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19021990

RESUMO

BACKGROUND: We sought to compare host immune cell kinetics, survival profile of donor skeletal myoblasts, and skeletal myoblast graft efficacy after autologous and allogeneic skeletal myoblast transplantation into a rat model of myocardial infarction. METHODS: One week after myocardial infarction, 128 animals were divided into four groups: group 1 (n = 24, receiving medium only), group 2 (n = 24, receiving medium and cyclosporine), group 3 (n = 40, autologous skeletal myoblast transplantation), and group 4 (n = 40, allogeneic skeletal myoblast transplantation with cyclosporine treatment). Rats were euthanized 10 minutes, 1 day, and 4, 7, and 28 days later. Host immune cell kinetics were assessed by immunohistochemical studies for macrophages, and CD4+ and CD8+ lymphocytes. Donor skeletal myoblast survival was confirmed by tracking prelabeled signals, and quantified by beta-gal assay. Heart function was evaluated by echocardiography. RESULTS: A transient immune cell infiltration was demonstrated in group 3, with macrophage infiltration on day 1 and day 4, CD8+ cell infiltration on day 4 and day 7, and CD4+ cell infiltration on day 4. In group 4, immunocyte infiltration was slightly more severe than that in group 3. Automyoblasts and allomyoblasts showed no significant difference of survival from day 1 to day 7 (p > 0.10); however, on day 28, automyoblasts showed better survival than allomyoblasts (p < 0.05). Transplantation of allomyoblasts increased systolic heart function and limited heart dilation after myocardial injury to a similar degree as automyoblasts (p > 0.10). CONCLUSIONS: The use of allomyoblasts is feasible and effective for cardiac repair with immunosuppressive treatment as compared with automyoblasts.


Assuntos
Transplante de Células/métodos , Ciclosporinas/farmacologia , Mioblastos Esqueléticos/transplante , Infarto do Miocárdio/cirurgia , Animais , Modelos Animais de Doenças , Feminino , Rejeição de Enxerto , Sobrevivência de Enxerto , Imuno-Histoquímica , Imunossupressores/farmacologia , Masculino , Mioblastos Esqueléticos/imunologia , Infarto do Miocárdio/patologia , Probabilidade , Distribuição Aleatória , Ratos , Ratos Wistar , Regeneração/fisiologia , Fatores de Risco , Sensibilidade e Especificidade , Transplante Autólogo , Transplante Homólogo , Remodelação Ventricular/fisiologia
6.
J Mol Cell Cardiol ; 45(1): 70-80, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18514708

RESUMO

We hypothesize that combining angiopoietin-1 (ANG-1) or ANG-2 with vascular endothelial growth factor (VEGF) improves myocardial perfusion and contractile function by modulating vascular adaptation of neoangiogenic microvessels in a chronic ischemic swine model. Four weeks after occlusion of the left circumflex coronary artery (LCx), animals were injected with AdVEGF(165) (n=6), AdVEGF(165)+AdANG-1 (n=6), AdVEGF(165)+AdANG-2 (n=6) or control vector (n=5) into the left ventricular posterolateral wall. Regional perfusion by fluorescent microspheres and segmental myocardial tissue velocity by tissue Doppler imaging (TDI) were assessed at baseline, 4 weeks post occlusion and 4 weeks post therapy. Despite similar vascular growth following VEGF+ANG-1 and VEGF+ANG-2 treatments, transmural myocardial contractility improved only when VEGF was paired with ANG-1. In contrast, regional systolic function deteriorated uniformly across subepicardial, mid-myocardial and subendocardial segments in VEGF and VEGF+ANG-2 treated groups. Contractile improvement was associated with enhanced vascular stability through augmented arteriole formation, tight structural integration between VE-cadherin and beta-catenin at endothelial junctions and improved cross-talk between endothelium and myocardium. Structural stability of developing intramyocardial microvessels contributes to systolic function during ischemic neovascularization. Coordinated regulation of angiogenic revascularization that supports vascular stability is a key aspect in improving therapeutic outcomes in ischemic myocardium.


Assuntos
Adenoviridae , Angiopoietina-1/biossíntese , Angiopoietina-2/biossíntese , Isquemia Miocárdica/metabolismo , Neovascularização Fisiológica , Recuperação de Função Fisiológica , Fator A de Crescimento do Endotélio Vascular/biossíntese , Angiopoietina-1/genética , Angiopoietina-2/genética , Animais , Antígenos CD/metabolismo , Arteríolas/metabolismo , Arteríolas/patologia , Caderinas/metabolismo , Doença Crônica , Circulação Coronária/genética , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Modelos Animais de Doenças , Ecocardiografia Doppler , Endotélio/metabolismo , Feminino , Terapia Genética/métodos , Masculino , Contração Miocárdica/genética , Isquemia Miocárdica/diagnóstico por imagem , Isquemia Miocárdica/patologia , Isquemia Miocárdica/terapia , Miocárdio/metabolismo , Miocárdio/patologia , Neovascularização Fisiológica/genética , Recuperação de Função Fisiológica/genética , Suínos , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/genética , beta Catenina/metabolismo
7.
J Thorac Cardiovasc Surg ; 134(5): 1332-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17976470

RESUMO

OBJECTIVE: We sought to investigate immune cell kinetics in relation to skeletal myoblast survival and heart function improvement after nonautologous skeletal myoblast transplantation in a rat model of myocardial infarction. METHODS: One week after myocardial infarction, 208 Wistar rats were grouped into group 1 (n = 24, receiving 150 muL of medium only), group 2 (n = 24, receiving 150 muL of medium and cyclosporine [INN: ciclosporin]), group 3 (n = 40, human skeletal myoblast transplantation), group 4 (n = 40, human skeletal myoblast transplantation with cyclosporine treatment), group 5 (n = 40, rat skeletal myoblast transplantation), and group 6 (n = 40, rat skeletal myoblast transplantation with cyclosporine treatment). The hearts were harvested at 10 minutes and 1, 4, 7, and 28 days after cell transplantation. Skeletal myoblast survival was confirmed by means of immunohistochemical studies and quantified by using real-time polymerase chain reaction. Host immune responses were assessed by immunostaining for macrophages and CD4+ and CD8+ lymphocytes. Heart function was evaluated by means of echocardiographic analysis. RESULTS: The majority of macrophages and lymphocytes infiltrated in the acute phase (from day 1 to day 7) and then subsided by day 28. The donor skeletal myoblasts survived and differentiated well in all skeletal myoblast transplantation groups. Allogeneic skeletal myoblasts showed a superior survival rate than xenogeneic skeletal myoblasts (P < .01). Cyclosporine inhibited the infiltration of the immunocytes, enhanced skeletal myoblast survival, and improved heart performance compared with that seen in the groups not receiving cyclosporine treatment (P < .05). CONCLUSIONS: Allomyoblasts survive better than do xenomyoblasts after transplantation into infarcted myocardium. After inhibition of immunocyte infiltration by means of immunosuppressive treatment, skeletal myoblast survival is enhanced, with improved heart performance. These findings suggest the feasibility of nonautologous myoblast transplantation with immunosuppressive treatment.


Assuntos
Transplante de Células , Coração/fisiopatologia , Mioblastos/transplante , Infarto do Miocárdio/terapia , Animais , Ciclosporina/farmacologia , Modelos Animais de Doenças , Feminino , Humanos , Tolerância Imunológica/efeitos dos fármacos , Imunossupressores/farmacologia , Masculino , Mioblastos/imunologia , Ratos , Ratos Wistar , Transplante Heterólogo/imunologia , Transplante Homólogo/imunologia
8.
Mol Cancer Res ; 5(7): 655-65, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17634421

RESUMO

Angiopoietins (ANG-1 and ANG-2) and their TIE-2 receptor tyrosine kinase have wide-ranging effects on tumor malignancy that includes angiogenesis, inflammation, and vascular extravasation. These multifaceted pathways present a valuable opportunity in developing novel inhibition strategies for cancer treatment. However, the regulatory role of ANG-1 and ANG-2 in tumor angiogenesis remains controversial. There is a complex interplay between complementary yet conflicting roles of both the ANGs in shaping the outcome of angiogenesis. Embryonic vascular development suggests that ANG-1 is crucial in engaging interaction between endothelial and perivascular cells. However, recruitment of perivascular cells by ANG-1 has recently been implicated in its antiangiogenic effect on tumor growth. It is becoming clear that TIE-2 signaling may function in a paracrine and autocrine manner directly on tumor cells because the receptor has been increasingly found in tumor cells. In addition, alpha(5)beta(1) and alpha(v)beta(5) integrins were recently recognized as functional receptors for ANG-1 and ANG-2. Therefore, both the ligands may have wide-ranging functions in cellular activities that affect overall tumor development. Collectively, these TIE-2-dependent and TIE-2-independent activities may account for the conflicting findings of ANG-1 and ANG-2 in tumor angiogenesis. These uncertainties have impeded development of a clear strategy to target this important angiogenic pathway. A better understanding of the molecular basis of ANG-1 and ANG-2 activity in the pathophysiologic regulation of angiogenesis may set the stage for novel therapy targeting this pathway.


Assuntos
Angiopoietinas/metabolismo , Neoplasias/irrigação sanguínea , Neovascularização Patológica/patologia , Receptores de TIE/metabolismo , Angiopoietinas/química , Animais , Humanos , Receptores de TIE/química
9.
J Biomed Sci ; 13(4): 579-91, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16547766

RESUMO

This study investigates the long-term angiogenic effects of ANG-1 and VEGF in a swine chronic myocardial ischemia model. Four-weeks after gradual occlusion of the left circumflex coronary artery by ameroid constrictor, animals were injected with recombinant adenoviral vectors carrying either human ANG-1 (n=9), human VEGF(165) (n=10) or empty vector (n=7) into the left ventricle free wall supplied by the constricted artery. Left ventricular perfusion in animals that received AdANG-1 (3.25+/-0.16 ml/min/g, p<0.05) recovered robustly 4 weeks after gene transfer while ischemia persisted in the AdVEGF (1.09+/-0.13 ml/min/g) and empty vector (1.20+/-0.03 ml/min/g) groups. Microvascular densities in the left ventricles of animals that received AdANG-1 (19.61+/-1.76/0.572 mm(2) myocardial tissue, p<0.05) and AdVEGF (18.17+/-1.43/0.572 mm(2) myocardial tissue, p<0.05) were significantly higher than animals that received empty vector (13.53+/-0.92/0.572 mm(2) myocardial tissue) 12 weeks after gene transfer. ANG-1, but not VEGF, contributed to enhanced regional perfusion by increasing arteriolar density (1.9+/-0.4/0.572 mm(2) myocardial tissue vs. 0.7+/-0.2/0.572 mm(2) myocardial tissue, p<0.05) of large-sized (50-100 microm) arterioles. These data demonstrate that gene transfer of ANG-1 and VEGF enhances angiogenesis, but ANG-1 promotes sustained improvement of ventricular perfusion that expedites recovery of ischemic myocardium via arteriogenesis.


Assuntos
Angiopoietina-1/farmacologia , Terapia Genética/métodos , Vetores Genéticos/farmacologia , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/terapia , Reperfusão Miocárdica , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Adenoviridae , Análise de Variância , Angiopoietina-1/genética , Animais , Angiografia Coronária , Circulação Coronária , Vasos Coronários/anatomia & histologia , Vasos Coronários/efeitos dos fármacos , Primers do DNA , Vetores Genéticos/genética , Humanos , Imuno-Histoquímica , Fluxo Sanguíneo Regional , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Fator A de Crescimento do Endotélio Vascular/genética
10.
Biochem Biophys Res Commun ; 324(2): 481-8, 2004 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-15474453

RESUMO

Bone marrow mesenchymal stem cells have been shown to transdifferentiate into cardiomyocytes after 5-azacytidine treatment or co-culturing with rodent cardiomyocytes. We investigate if adult human bone marrow stem cells can be differentiated ex vivo into cardiomyocyte-like cells (CLCs) independent of cytotoxic agents or co-culturing technique. Sternal bone marrow was collected from 16 patients undergoing coronary artery bypass surgery. Mesenchymal stem cells were differentiated in a cardiomyogenic differentiation medium containing insulin, dexamethasone, and ascorbic acid. Differentiation towards CLCs was determined by induced expression of cardiomyocyte-specific proteins. Differentiated CLCs expressed multiple structural and contractile proteins that are associated with cardiomyocytes. Thin filament associated myofibrillar proteins were detected early in the cells, with cardiac troponin I, sarcomeric tropomyosin, and cardiac titin among the first expressed. Some CLCs were found to develop into a nascent cardiomyocyte phenotype with cross-striated myofibrils characterized by alpha-actinin-positive Z bands after 4-5 passages in differentiated culture. These lineage-defined CLCs may be potentially useful for repairing damaged myocardium.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células/métodos , Miócitos Cardíacos/metabolismo , Adipócitos/metabolismo , Idoso , Ácido Ascórbico/farmacologia , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Meios de Cultura/farmacologia , Primers do DNA/química , Dexametasona/farmacologia , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Mesoderma/citologia , Pessoa de Meia-Idade , Músculo Esquelético/citologia , Miocárdio/metabolismo , Fenótipo , RNA/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Fatores de Tempo
11.
J Card Surg ; 19(2): 161-6, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15016059

RESUMO

BACKGROUND: Transmyocardial laser revascularization (TMLR) has been shown to induce angiogenesis and improve regional myocardial blood flow. However, the biological response to laser of different energy doses in nonischemic myocardium has not been well studied. We study the time course of histological appearance following high- and low-energy TMLR in a rodent model of normal myocardium. METHODS: The animals were randomized to two groups (high-energy (1.4 J/pulse) TMLR group and low-energy (0.6 J/ pulse) TMLR group). TMLR was applied to the anterolateral wall of the left ventricle. Then the animals were sacrificed (one every 2 or 3 days up to 6 weeks, then one every week) to give a series of hearts from 1 week to 12 weeks following TMLR. RESULTS: No patent laser channels were identified. All laser channel remnants were composed of granulation tissue, fibrosis, and various amounts of vascular structures. Changes in vascular density demonstrated a similar pattern in both groups over time. The initial increase in vascular density within the laser channel remnants and the surrounding areas was more marked in group I than in group II. Gradual decline of vascular density occurred during a later follow-up. More fibrosis and less vasculature were demonstrated 12 weeks after laser therapy in group I. CONCLUSION: Angiogenic response to the Holmium:YAG laser in the nonischemic myocardium is nonspecific and unsustainable. TMLR might chronically impair the myocardium function by enhancing regional scar formation. Therefore, we should clearly identify the ischemic area for laser therapy. Reduction of laser energy might prevent this potential complication.


Assuntos
Terapia a Laser , Isquemia Miocárdica/cirurgia , Revascularização Miocárdica , Miocárdio/patologia , Animais , Modelos Animais de Doenças , Fibrose , Seguimentos , Modelos Cardiovasculares , Isquemia Miocárdica/fisiopatologia , Ratos , Ratos Wistar , Fatores de Tempo , Grau de Desobstrução Vascular/fisiologia
12.
Exp Cell Res ; 279(2): 299-309, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12243755

RESUMO

Angiopoietins have been increasingly implicated to play important roles in blood vessel formation, remodeling, maturation, and maintenance. However, their roles in tumor angiogenesis and hence tumor growth and metastasis still remain uncertain. In this work, angiopoietin 1 expression was amplified in human cervical cancer HeLa cells by stable transfection or recombinant human adenovirus-mediated gene transfer. We show that increased angiopoietin 1 expression promoted in vivo growth of human cervical cancers in mice by promoting tumor angiogenesis and inhibiting tumor cell apoptosis. Furthermore, we also show for the first time that overexpression of angiopoietin 1 also leads to increased tumor vessel plasticity with a large number of vessels lacking periendothelial supporting cells. These results indicate that angiopoietin 1 promotes tumor angiogenesis and tumor vessel plasticity of human cervical cancer in mice.


Assuntos
Adenocarcinoma/metabolismo , Vasos Sanguíneos/metabolismo , Glicoproteínas de Membrana/metabolismo , Neovascularização Patológica/fisiopatologia , Neoplasias do Colo do Útero/metabolismo , Adenocarcinoma/patologia , Adenoviridae/genética , Adenoviridae/metabolismo , Angiopoietina-1 , Animais , Fatores de Crescimento Endotelial/metabolismo , Feminino , Técnicas de Transferência de Genes , Células HeLa , Humanos , Linfocinas/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Neoplasias do Colo do Útero/patologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
13.
J Card Surg ; 17(4): 350-4, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12546086

RESUMO

Therapeutic angiogenesis may be a realistic approach in treating ischemic heart disease. VEGF is a major angiogenic factor involved in physiological as well as pathological angiogenesis. The ability of VEGF to promote angiogenesis in animal and clinical studies has been studied extensively. However, it is becoming clear that VEGF alone may not be sufficient to effectively complete the angiogenesis process. The use of more than one growth factor may be more pertinent in creating a sustainable angiogenic effect with clinically significant outcome. The challenge is to find complementary partners in angiogenesis to better affect the outcome of the process. To this end, we have been studying the effects of other angiogenic factors such as angiopoietin-1 (Ang-1) in a chronic ischemic porcine model. Single intramyocardial introduction of adenovirus-mediated gene transfer of Ang-1 into the left ventricle free wall has been found to enhance angiogenesis by augmenting the formation of new capillaries that manifested in improved total blood flow in the myocardium. A combined therapeutic angiogenesis study involving VEGF and Ang-1 is currently underway. Due to their unique complementary properties, it is expected that the combination will not merely enhance angiogenesis but will also lead to healthy and mature vascular network in the ischemic myocardium.


Assuntos
Doença da Artéria Coronariana/terapia , Neovascularização Patológica/terapia , Indutores da Angiogênese/uso terapêutico , Angioplastia Coronária com Balão , Angiopoietina-1 , Animais , Ponte de Artéria Coronária , Fatores de Crescimento Endotelial/uso terapêutico , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Linfocinas/uso terapêutico , Glicoproteínas de Membrana/uso terapêutico , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA