Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Transpl Int ; 26(11): 1126-37, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24102821

RESUMO

Heart transplant gene therapy requires vectors with long-lasting gene expression, high cardiotropism, and minimal pathological effects. Here, we examined transduction properties of ex vivo intracoronary delivery of adeno-associated virus (AAV) serotype 2, 8, and 9 in rat syngenic and allogenic heart transplants. Adult Dark Agouti (DA) rat hearts were intracoronarily perfused ex vivo with AAV2, AAV8, or AAV9 encoding firefly luciferase and transplanted heterotopically into the abdomen of syngenic DA or allogenic Wistar-Furth (WF) recipients. Serial in vivo bioluminescent imaging of syngraft and allograft recipients was performed for 6 months and 4 weeks, respectively. Grafts were removed for PCR-, RT-PCR, and luminometer analysis. In vivo bioluminescent imaging of recipients showed that AAV9 induced a prominent and stable luciferase activity in the abdomen, when compared with AAV2 and AAV8. However, ex vivo analyses revealed that intracoronary perfusion with AAV2 resulted in the highest heart transplant transduction levels in syngrafts and allografts. Ex vivo intracoronary delivery of AAV2 resulted in efficient transgene expression in heart transplants, whereas intracoronary AAV9 escapes into adjacent tissues. In terms of cardiac transduction, these results suggest AAV2 as a potential vector for gene therapy in preclinical heart transplants studies, and highlight the importance of delivery route in gene transfer studies.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Transplante de Coração , Coração/virologia , Animais , Dependovirus/classificação , Isoenxertos , Luciferases/genética , Masculino , Miocárdio/metabolismo , Ratos , Transgenes/genética
2.
J Heart Lung Transplant ; 29(9): 1058-66, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20580263

RESUMO

BACKGROUND: Hypoxia-inducible factor-1 (HIF-1), a key transcription factor in hypoxia, affects a wide range of adaptive cell functions. We examined the kinetics of endogenous HIF-1alpha during acute and chronic rejection, and the effect of exogenous HIF-1alpha in chronically rejecting rat cardiac allografts. METHODS: Heterotopic cardiac transplantations were performed between major MHC-mismatched Dark Agouti and Wistar-Furth rats. Cyclosporine A (CsA) was used to prevent acute rejection in the chronic rejection model. The effect of HIF-1alpha overexpression was investigated by adeno-assocated virus 2 (AAV2)-mediated gene transfer of a constitutively stabilized form of mouse HIF-1alpha (AAV-HIF-1alpha). The analysis of allografts was based on histology, immunohistochemistry and quantitative reverse transcript-polymerase chain reaction (RT-PCR). RESULTS: Acute and chronic rejection significantly induced HIF-1alpha mRNA in rat cardiac allografts when compared with syngeneic controls. Immunohistochemistry localized significantly increased HIF-1alpha immunoreactivity to vascular smooth muscle cells, vascular endothelial cells, post-capillary venules and graft-infiltrating mononuclear inflammatory cells of the allograft, whereas expression in cardiomyocytes remained unchanged. Regression analysis revealed a linear correlation between the progression of cardiac allograft vasculopathy (CAV) and HIF-1alpha immunoreactivity in post-capillary venules and graft-infiltrating mononuclear inflammatory cells in chronically rejecting rat cardiac allografts. AAV-HIF-1alpha enhanced cardiomyocyte HIF-1alpha production and significantly reduced cardiomyocyte apoptosis and the development of CAV in chronically rejecting rat cardiac allografts. CONCLUSIONS: We found that acute and chronic rejection increased HIF-1alpha mRNA and protein levels in rat cardiac allografts. On the other hand, cardiomyocyte-targeted HIF-1alpha gene transfer inhibited cardiomyocyte apoptosis and the development of CAV, suggesting a novel therapeutic strategy for HIF-1alpha in cardiac allografts.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Transplante de Coração/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Abdome , Animais , Apoptose , Anidrase Carbônica IX , Anidrases Carbônicas/metabolismo , Rejeição de Enxerto/patologia , Transplante de Coração/imunologia , Transplante de Coração/métodos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Complicações Pós-Operatórias/patologia , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos , Ratos Endogâmicos WF , Transplante Heterotópico , Transplante Homólogo/imunologia , Transplante Homólogo/patologia
3.
J Neurochem ; 112(6): 1513-26, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20050971

RESUMO

Previous studies have shown that the maintenance of post-mitotic state is critical for the life-long survival of the inner ear mechanosensory cells, the hair cells. A general concept is that differentiated, post-mitotic cells rapidly die following cell cycle re-entry. Here we have compared the response of postnatal cochlear (auditory) and utricular (balance) hair cells to forced cell cycle reactivation and p53 up-regulation. Forced S-phase entry was triggered through the human papillomavirus-16 E7 oncogene misexpression in explant cultures. It induced DNA damage and p53 induction in cochlear outer hair cells and these cells were rapidly lost, before entry into mitosis. The death was attenuated by p53 inactivation. In contrast, despite DNA damage and p53 induction, utricular hair cells showed longer term survival and a proportion of them progressed into mitosis. Consistently, pharmacological elevation of p53 levels by nutlin-3a led to a death-prone phenotype of cochlear outer hair cells, while other hair cell populations were death-resistant. These data have important clinical implications as they show the importance of p53 in sensory cells that are essential in hearing function.


Assuntos
Ciclo Celular/fisiologia , Orelha Interna/citologia , Mecanorreceptores/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/fisiologia , Adenoviridae/fisiologia , Animais , Animais Recém-Nascidos , Calbindinas , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Dano ao DNA/fisiologia , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Proteínas de Fluorescência Verde/genética , Imidazóis/farmacologia , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Parvalbuminas/genética , Parvalbuminas/metabolismo , Piperazinas/farmacologia , Proteína G de Ligação ao Cálcio S100/genética , Proteína G de Ligação ao Cálcio S100/metabolismo , Fatores de Tempo , Proteína Supressora de Tumor p53/deficiência , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Tropismo Viral/genética , Tropismo Viral/fisiologia
4.
Arterioscler Thromb Vasc Biol ; 29(5): 691-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19213942

RESUMO

OBJECTIVE: Chronic rejection is the main reason for the poor long-term survival of heart transplant recipients and is characterized by cardiac allograft inflammation, fibrosis, and arteriosclerosis. We examined the specific roles of different platelet-derived growth factor (PDGF) ligands (A-D)--potent mesenchymal cell mitogens--in rat cardiac allografts. METHODS AND RESULTS: PDGFR-alpha mRNA was upregulated in acutely-rejecting, and PDGF-A and PDGF-C mRNA in chronically-rejecting cardiac centhatn allografts. In acute rejection, PDGFR-alpha immunoreactivity increased in the media of arteries. In chronically-rejecting allografts, immunoreactivity of all PDGF ligands and receptors--except that of PDGF-B ligand--was found in the intima of arteries, and the expression of PDGF-A and PDGF-C was seen in cardiomyocytes. Intracoronary adeno-associated virus-2 (AAV2)-mediated PDGF-A and -D gene transfer enhanced cardiac allograft inflammation. AAV2-PDGF-A, AAV2-PDGF-C, and AAV2-PDGF-D significantly upregulated profibrotic TGF-beta1 mRNA and accelerated cardiac fibrosis and arteriosclerosis. In contrast, AAV2-PDGF-B did not aggravate chronic rejection. CONCLUSIONS: We found that alloimmune response induces PDGF-A, PDGF-C, and PDGF-D expression in the graft vasculature. PDGF-A, PDGF-C, and PDGF-D mediated profibrotic and proarteriosclerotic effects in transplanted hearts involving the TGF-beta1 pathway. Inhibition of signaling of all PDGF-ligands except that of PDGF-B may thus be needed to inhibit chronic rejection in cardiac allografts.


Assuntos
Arteriosclerose/imunologia , Rejeição de Enxerto/metabolismo , Transplante de Coração/imunologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Modelos Animais de Doenças , Fibrose/imunologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , RNA Mensageiro/metabolismo , Ratos , Transdução de Sinais , Transplante Homólogo , Regulação para Cima
5.
Circ Res ; 100(10): 1468-75, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17478733

RESUMO

Vascular endothelial growth factor (VEGF)-C and VEGF-D are composed of the receptor-binding VEGF homology domain and a carboxy-terminal silk homology domain that requires proteolytic cleavage for growth factor activation. Here, we explored whether the C-terminal heparin-binding domain of the VEGF(165) or VEGF(189) isoform also containing neuropilin-binding sequences could substitute for the silk homology domain of VEGF-C. Such VEGF-C/VEGF-heparin-binding domain chimeras were produced and shown to activate VEGF-C receptors, and, when expressed in tissues via adenovirus or adeno-associated virus vectors, stimulated lymphangiogenesis in vivo. However, both chimeras induced a distinctly different pattern of lymphatic vessels when compared with VEGF-C. Whereas VEGF-C-induced vessels were initially a dense network of small diameter vessels, the lymphatic vessels induced by the chimeric growth factors tended to form directly along tissue borders, along basement membranes that are rich in heparan sulfate. For example, in skeletal muscle, the chimeras induced formation of lumenized lymphatic vessels more efficiently than wild-type VEGF-C. We conclude that the matrix-binding domain of VEGF can target VEGF-C activity to heparin-rich basement membrane structures. These properties may prove useful for tissue engineering and attempts to regenerate lymphatic vessels in lymphedema patients.


Assuntos
Heparina/metabolismo , Vasos Linfáticos/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator C de Crescimento do Endotélio Vascular/farmacologia , Adenoviridae/genética , Animais , Sítios de Ligação , Células Cultivadas , Humanos , Linfangiogênese/efeitos dos fármacos , Camundongos , Proteínas Recombinantes de Fusão/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Circ Res ; 98(11): 1373-80, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16690881

RESUMO

Angiopoietin-1 (Ang1) and Ang2 regulate the maintenance of normal vasculature by direct endothelial and indirect smooth muscle cell (SMC) effects. Dysfunction of vascular wall cells is considered central in cardiac allograft vasculopathy (CAV), where inflammation and arterial injury initiate subsequent intimal SMC proliferation. In this study, we investigated the effect of exogenous Ang1 and Ang2 in chronically rejecting rat cardiac allografts by intracoronary adeno-associated virus (AAV)-mediated gene transfer. Bioluminescent imaging of AAV-transfected syngeneic grafts revealed gradual and stable transgene expression in graft cardiomyocytes. In cardiac allografts, both AAV-Ang1 and AAV-Ang2 decreased inflammation and increased antiapoptotic Bcl-2 mRNA and Bcl-2/Bax ratio at 8 weeks. Only AAV-Ang2 decreased the development of CAV, whereas AAV-Ang1 activated arterial SMC and increased PDGF-A mRNA in the allograft. Collectively, our results show that exogenous Ang1 and Ang2 have similar antiinflammatory and antiapoptotic effects in cardiac allografts. Prolonged AAV-mediated Ang1 transgene expression also induced SMC activation, whereas AAV-Ang2 lacked the SMC activating effects and decreased CAV. Our results thus highlight the common protective and diverse SMC effects of Ang1 and Ang2 in cardiac allograft microenvironment and the importance of timing of angiopoietins to achieve therapeutic effects.


Assuntos
Angiopoietina-1/farmacologia , Angiopoietina-2/farmacologia , Transplante de Coração/efeitos adversos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Doenças Vasculares/etiologia , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Angiopoietina-2/genética , Angiopoietina-2/metabolismo , Animais , Apoptose/genética , Proliferação de Células/efeitos dos fármacos , Dependovirus/genética , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Rejeição de Enxerto/fisiopatologia , Substâncias de Crescimento/genética , Humanos , Masculino , Músculo Liso Vascular/fisiopatologia , Miocardite/etiologia , Miocardite/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos de Músculo Liso/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos , Doenças Vasculares/patologia , Doenças Vasculares/fisiopatologia , Doenças Vasculares/prevenção & controle , Proteína X Associada a bcl-2/metabolismo
7.
Cancer Res ; 65(11): 4739-46, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15930292

RESUMO

Lymphangiogenic growth factors vascular endothelial growth factor (VEGF)-C and VEGF-D have been shown to promote lymphatic metastasis by inducing tumor-associated lymphangiogenesis. In this study, we have investigated how tumor cells gain access into lymphatic vessels and at what stage tumor cells initiate metastasis. We show that VEGF-C produced by tumor cells induced extensive lymphatic sprouting towards the tumor cells as well as dilation of the draining lymphatic vessels, suggesting an active role of lymphatic endothelial cells in lymphatic metastasis. A significant increase in lymphatic vessel growth occurred between 2 and 3 weeks after tumor xenotransplantation, and lymph node metastasis occurred at the same stage. These processes were blocked dose-dependently by inhibition of VEGF receptor 3 (VEGFR-3) signaling by systemic delivery of a soluble VEGFR-3-immunoglobulin (Ig) fusion protein via adenoviral or adeno-associated viral vectors. However, VEGFR-3-Ig did not suppress lymph node metastasis when the treatment was started at a later stage after the tumor cells had already spread out, suggesting that tumor cell entry into lymphatic vessels is a key step during tumor dissemination via the lymphatics. Whereas lymphangiogenesis and lymph node metastasis were significantly inhibited by VEGFR-3-Ig, some tumor cells were still detected in the lymph nodes in some of the treated mice. This indicates that complete blockade of lymphatic metastasis may require the targeting of both tumor lymphangiogenesis and tumor cell invasion.


Assuntos
Endotélio Linfático/patologia , Neoplasias Pulmonares/patologia , Linfonodos/patologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Imunoglobulinas/administração & dosagem , Imunoglobulinas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Metástase Linfática , Camundongos , Camundongos SCID , Estadiamento de Neoplasias , Neovascularização Patológica/patologia , Neovascularização Patológica/prevenção & controle , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais/fisiologia , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/administração & dosagem , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
FASEB J ; 19(10): 1365-7, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15958522

RESUMO

Therapeutic angiogenesis provides a potential alternative for the treatment of cardiovascular ischemic diseases. Vascular endothelial growth factor (VEGF) is an important component of the angiogenic response to ischemia. Here we used adeno-associated virus (AAV) gene delivery to skeletal muscle to examine the effects of VEGF vs. a stabilized form of hypoxia-inducible factor-1alpha (HIF-1alpha). The recombinant AAVs were injected into mouse tibialis anterior muscle, and their effects were analyzed by immunohistochemistry and functional assays. These analyses showed that stabilized HIF-1alpha markedly increase capillary sprouting and proliferation, whereas VEGF164 or VEGF120 induced only proliferation of endothelial cells without formation of proper capillary structures. The Evans Blue permeability assay indicated that, unlike VEGF, HIF-1alpha overexpression did not increase vascular leakiness in the transduced muscle. Doppler ultrasound imaging showed that vascular perfusion in the HIF-1alpha treated muscles was significantly enhanced when compared to the controls and not further improved by co-expression of the arteriogenic growth factors angiopoietin-1 or platelet-derived growth factor-B. Our results show that AAV-mediated transduction of a stabilized form of HIF-1alpha can circumvent the problems associated with overexpression of individual angiogenic growth factors. HIF-1alpha should thus offer a potent alternative for pro-angiogenic gene therapy.


Assuntos
Dependovirus/genética , Transferência Genética Horizontal , Terapia Genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Permeabilidade Capilar , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Perfusão , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Proteínas Proto-Oncogênicas c-sis/genética
9.
Blood ; 105(12): 4642-8, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15746084

RESUMO

Angiopoietin 1 (Ang1), a ligand for the receptor tyrosine kinase Tie2, regulates the formation and stabilization of the blood vessel network during embryogenesis. In adults, Ang1 is associated with blood vessel stabilization and recruitment of perivascular cells, whereas Ang2 acts to counter these actions. Recent results from gene-targeted mice have shown that Ang2 is also essential for the proper patterning of lymphatic vessels and that Ang1 can be substituted for this function. In order to characterize the effects of the angiopoietins on lymphatic vessels, we employed viral vectors for overexpression of Ang1 in adult mouse tissues. We found that Ang1 activated lymphatic vessel endothelial proliferation, vessel enlargement, and generation of long endothelial cell filopodia that eventually fused, leading to new sprouts and vessel development. Cutaneous lymphatic hyperplasia was also detected in transgenic mice expressing Ang1 in the basal epidermal cells. Tie2 was expressed in the lymphatic endothelial cells and Ang1 stimulation of these cells resulted in up-regulation of vascular endothelial growth factor receptor 3 (VEGFR-3). Furthermore, a soluble form of VEGFR-3 inhibited the observed lymphatic sprouting. Our results reinforce the concept that Ang1 therapy may be useful in settings of tissue edema.


Assuntos
Angiopoietina-1/fisiologia , Endotélio Vascular/citologia , Hiperplasia/patologia , Sistema Linfático/fisiologia , Neovascularização Fisiológica , Adenoviridae/genética , Animais , Northern Blotting , Proliferação de Células , Células Cultivadas , Clonagem Molecular , Derme/metabolismo , Edema , Endotélio/citologia , Células Epidérmicas , Vetores Genéticos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Estrutura Terciária de Proteína , Receptor TIE-2/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Regulação para Cima , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
Blood ; 104(10): 3198-204, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15271796

RESUMO

Platelet-derived growth factor-D (PDGF-D) is a recently characterized member of the PDGF family with unknown in vivo functions. We investigated the effects of PDGF-D in transgenic mice by expressing it in basal epidermal cells and then analyzed skin histology, interstitial fluid pressure, and wound healing. When compared with control mice, PDGF-D transgenic mice displayed increased numbers of macrophages and elevated interstitial fluid pressure in the dermis. Wound healing in the transgenic mice was characterized by increased cell density and enhanced recruitment of macrophages. Macrophage recruitment was also the characteristic response when PDGF-D was expressed in skeletal muscle or ear by an adeno-associated virus vector. Combined expression of PDGF-D with vascular endothelial growth factor-E (VEGF-E) led to increased pericyte/smooth muscle cell coating of the VEGF-E-induced vessels and inhibition of the vascular leakiness that accompanies VEGF-E-induced angiogenesis. These results show that full-length PDGF-D is activated in tissues and is capable of increasing interstitial fluid pressure and macrophage recruitment and the maturation of blood vessels in angiogenic processes.


Assuntos
Linfocinas/genética , Linfocinas/metabolismo , Macrófagos/fisiologia , Neovascularização Fisiológica/fisiologia , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Vasos Sanguíneos/fisiologia , Movimento Celular/imunologia , Derme/fisiologia , Líquido Extracelular/fisiologia , Humanos , Macrófagos/citologia , Camundongos , Camundongos Transgênicos , Músculo Esquelético/fisiologia , Pressão , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Virais/genética , Cicatrização
11.
J Gene Med ; 6(5): 545-54, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15133765

RESUMO

BACKGROUND: Gene transfer offers considerable potential for altering vessel wall physiology and intervention in vascular disease. Therefore, there is great interest in developing optimal strategies and vectors for efficient, targeted gene delivery into a vessel wall. METHODS: We studied adeno-associated viruses (AAV; 9 x 10(8) to 4 x 10(9) TU/ml) for their usefulness to transduce rabbit arteries in vivo in comparison with adenoviruses (Adv; 1 x 10(9) to 1 x 10(10) pfu/ml). 100 microl of viruses or placebo solution were injected intraluminally into transiently isolated carotid segments. RESULTS: In normal arteries AAV transduced mainly medial smooth muscle cells (SMC) while Adv transduced exclusively endothelial cells (EC). Mechanical injury to EC layer and internal elastic lamina enabled Adv to penetrate and transduce medial SMC. Transgene expression in EC after the AAV-mediated gene transfer was very low. The use of the EC-specific Tie-1 promoter did not lead to specific transgene expression in EC. Transgene expression in SMC persisted for at least 100 days after the AAV treatment whereas the Adv-mediated effect diminished in 14 days. AAV caused only a modest increase in EC VCAM-1 expression and proliferation rate of vascular cells as compared with the mock-treated arteries while Adv caused an extensive inflammatory cell infiltration, VCAM-1 expression, vascular cell proliferation and morphological damages. CONCLUSIONS: Significant differences were observed between the AAV and the Adv vectors in their patterns of arterial transduction and consequent inflammatory responses. These distinct properties may be utilized for different applications in vascular biology research and gene therapy for cardiovascular diseases.


Assuntos
Adenoviridae/genética , Artérias Carótidas/metabolismo , Dependovirus/genética , Endotélio Vascular/metabolismo , Técnicas de Transferência de Genes , Músculo Liso Vascular/metabolismo , Animais , Artérias Carótidas/patologia , Proliferação de Células , Endotélio Vascular/citologia , Vetores Genéticos/efeitos adversos , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Inflamação/etiologia , Inflamação/patologia , Leucócitos/patologia , Masculino , Músculo Liso Vascular/citologia , Regiões Promotoras Genéticas , Coelhos , Receptor de TIE-1/genética , Transdução Genética , Molécula 1 de Adesão de Célula Vascular/biossíntese
12.
Mol Ther ; 8(6): 903-10, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14664792

RESUMO

Macrophage scavenger receptors (MSR) promote atherosclerotic lesion formation, and modulation of MSR activity has been shown to influence atherosclerosis. Soluble receptors are effective in inhibiting receptor-mediated functions in various diseases. We have generated a secreted macrophage scavenger receptor (sMSR) that consists of the bovine growth hormone signal sequence and the human MSR A I extracellular domains. sMSR reduces degradation of atherogenic modified low-density lipoproteins and monocyte/macrophage adhesion on endothelial cells in vitro. To test long-term effects of sMSR, atherosclerosis-susceptible LDLR knockout mice were transduced via the tail vein with an adeno-associated virus (AAV) expressing sMSR or control enhanced green fluorescent protein (EGFP), and a Western-type diet was started. Gene transfer caused a temporary elevation in alkaline phosphatase and aspartate amino transferase values without a change in C-reactive protein. sMSR protein was detected in the plasma of the transduced mice by a specific ELISA 6 months after the gene transfer. AAV-mediated sMSR gene transfer reduced atherosclerotic lesion area in the aorta by 21% (P < 0.05) compared to EGFP-transduced control mice. Even though eradication of established disease was not possible, atherosclerotic lesion formation could be modified using AAV-mediated gene transfer of the decoy sMSR.


Assuntos
Arteriosclerose/terapia , Dependovirus , Terapia Genética , Vetores Genéticos , Receptores Imunológicos/genética , Animais , Aorta/patologia , Arteriosclerose/patologia , Humanos , Lipídeos/sangue , Camundongos , Camundongos Knockout , Receptores Imunológicos/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores Depuradores
13.
J Virol ; 76(22): 11530-40, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388714

RESUMO

Adeno-associated viruses (AAVs) are promising vectors for various gene therapy applications due to their long-lasting transgene expression and wide spectrum of target cells. Recently, however, it has become apparent that there are considerable differences in the efficiencies of transduction of different cell types by AAVs. Here, we analyzed the efficiencies of transduction and the transport mechanisms of AAV type 2 (AAV-2) in different cell types, emphasizing endothelial cells. Expression analyses in both cultured cells and the rabbit carotid artery assay showed a remarkably low level of endothelial cell transduction in comparison to the highly permissive cell types. The study of the endosomal pathways of AAV-2 with fluorescently labeled virus showed clear targeting of the Golgi area in permissive cell lines, but this phenomenon was absent in the endothelial cell line EAhy-926. On the other hand, the response to the block of endosomal acidification by bafilomycin A1 also showed differences among the permissive cell types. We also analyzed the effect of proteasome inhibitors on endothelial cells, but their impact on the primary cells and in vivo was not significant. On the contrary, analysis of the expression pattern of heparan sulfate proteoglycans (HSPGs), the primary receptors of AAV-2, revealed massive deposits of HSPG in the extracellular matrix of endothelial cells. The matrix-associated receptors may therefore compete for virus binding and reduce transduction in endothelial cells. Accordingly, in endothelial cells detached from their matrix, AAV-2 transduction was significantly increased. Altogether, these results point to a more complex cell-type-specific mode of transduction of AAV-2 than previously appreciated.


Assuntos
Dependovirus/patogenicidade , Endotélio Vascular/virologia , Vetores Genéticos , Proteínas Luminescentes/metabolismo , Transdução Genética , Animais , Artérias Carótidas/citologia , Artérias Carótidas/virologia , Linhagem Celular , Células Cultivadas , Dependovirus/genética , Dependovirus/metabolismo , Endotélio Vascular/citologia , Citometria de Fluxo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/genética , Masculino , Coelhos
14.
J Exp Med ; 196(6): 719-30, 2002 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-12235206

RESUMO

Recent work from many laboratories has demonstrated that the vascular endothelial growth factor-C/VEGF-D/VEGFR-3 signaling pathway is crucial for lymphangiogenesis, and that mutations of the Vegfr3 gene are associated with hereditary lymphedema. Furthermore, VEGF-C gene transfer to the skin of mice with lymphedema induced a regeneration of the cutaneous lymphatic vessel network. However, as is the case with VEGF, high levels of VEGF-C cause blood vessel growth and leakiness, resulting in tissue edema. To avoid these blood vascular side effects of VEGF-C, we constructed a viral vector for a VEGFR-3-specific mutant form of VEGF-C (VEGF-C156S) for lymphedema gene therapy. We demonstrate that VEGF-C156S potently induces lymphangiogenesis in transgenic mouse embryos, and when applied via viral gene transfer, in normal and lymphedema mice. Importantly, adenoviral VEGF-C156S lacked the blood vascular side effects of VEGF and VEGF-C adenoviruses. In particular, in the lymphedema mice functional cutaneous lymphatic vessels of normal caliber and morphology were detected after long-term expression of VEGF-C156S via an adeno associated virus. These results have important implications for the development of gene therapy for human lymphedema.


Assuntos
Fatores de Crescimento Endotelial/genética , Terapia Genética , Sistema Linfático/fisiologia , Neovascularização Fisiológica , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Fatores de Crescimento/fisiologia , Adenoviridae/genética , Animais , Dependovirus/genética , Sistema Linfático/embriologia , Linfedema/terapia , Camundongos , RNA Mensageiro/análise , Fator C de Crescimento do Endotélio Vascular , Receptor 3 de Fatores de Crescimento do Endotélio Vascular
15.
FASEB J ; 16(9): 1041-9, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12087065

RESUMO

Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are important regulators of blood and lymphatic vessel growth and vascular permeability. The VEGF-C/VEGFR-3 signaling pathway is crucial for lymphangiogenesis, and heterozygous inactivating missense mutations of the VEGFR-3 gene are associated with hereditary lymphedema. However, VEGF-C can have potent effects on blood vessels because its receptor VEGFR-3 is expressed in certain blood vessels and because the fully processed form of VEGF-C also binds to the VEGFR-2 of blood vessels. To characterize the in vivo effects of VEGF-C on blood and lymphatic vessels, we have overexpressed VEGF-C via adenovirus- and adeno-associated virus-mediated transfection in the skin and respiratory tract of athymic nude mice. This resulted in dose-dependent enlargement and tortuosity of veins, which, along with the collecting lymphatic vessels were found to express VEGFR-2. Expression of angiopoietin 1 blocked the increased leakiness of the blood vessels induced by VEGF-C whereas vessel enlargement and lymphangiogenesis were not affected. However, angiogenic sprouting of new blood vessels was not observed in response to AdVEGF-C or AAV-VEGF-C. These results show that virally produced VEGF-C induces blood vessel changes, including vascular leak, but its angiogenic potency is much reduced compared with VEGF in normal skin.


Assuntos
Adenoviridae/genética , Fatores de Crescimento Endotelial/genética , Neovascularização Fisiológica , Pele/irrigação sanguínea , Angiopoietina-1 , Animais , Vasos Sanguíneos/anatomia & histologia , Vasos Sanguíneos/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Linhagem Celular , Dependovirus/genética , Fatores de Crescimento Endotelial/metabolismo , Vetores Genéticos , Sistema Linfático/crescimento & desenvolvimento , Linfocinas/genética , Glicoproteínas de Membrana/farmacologia , Camundongos , Camundongos Nus , Mucosa Nasal/irrigação sanguínea , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular , Pele/metabolismo , Transfecção , Fator A de Crescimento do Endotélio Vascular , Fator C de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA