Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 35(4): 948-59, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25810299

RESUMO

OBJECTIVE: Activation of liver X receptor (LXR) inhibits atherosclerosis but induces hypertriglyceridemia. In vitro, it has been shown that mitogen-activated protein kinase kinase 1/2 (MEK1/2) inhibitor synergizes LXR ligand-induced macrophage ABCA1 expression and cholesterol efflux. In this study, we determined whether MEK1/2 (U0126) and LXR ligand (T0901317) can have a synergistic effect on the reduction of atherosclerosis while eliminating LXR ligand-induced fatty livers and hypertriglyceridemia. We also set out to identify the cellular mechanisms of the actions. APPROACH AND RESULTS: Wild-type mice were used to determine the effect of U0126 on a high-fat diet or high-fat diet plus T0901317-induced transient dyslipidemia and liver injury. ApoE deficient (apoE(-/-)) mice or mice with advanced lesions were used to determine the effect of the combination of T0901317 and U0126 on atherosclerosis and hypertriglyceridemia. We found that U0126 protected animals against T0901317-induced transient or long-term hepatic lipid accumulation, liver injury, and hypertriglyceridemia. Meanwhile, the combination of T0901317 and U0126 inhibited the development of atherosclerosis in a synergistic manner and reduced advanced lesions. Mechanistically, in addition to synergistic induction of macrophage ABCA1 expression, the combination of U0126 and T0901317 maintained arterial wall integrity, inhibited macrophage accumulation in aortas and formation of macrophages/foam cells, and activated reverse cholesterol transport. The inhibition of T0901317-induced lipid accumulation by the combined U0126 might be attributed to inactivation of lipogenesis and activation of lipolysis/fatty acid oxidation pathways. CONCLUSIONS: Our study suggests that the combination of mitogen-activated protein kinase kinase 1/2 inhibitor and LXR ligand can function as a novel therapy to synergistically reduce atherosclerosis while eliminating LXR-induced deleterious effects.


Assuntos
Doenças da Aorta/prevenção & controle , Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Butadienos/farmacologia , Hidrocarbonetos Fluorados/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Nitrilas/farmacologia , Receptores Nucleares Órfãos/agonistas , Inibidores de Proteínas Quinases/farmacologia , Sulfonamidas/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/enzimologia , Aorta/patologia , Doenças da Aorta/enzimologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/enzimologia , Aterosclerose/genética , Aterosclerose/patologia , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Colesterol/metabolismo , Modelos Animais de Doenças , Sinergismo Farmacológico , Quimioterapia Combinada , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/enzimologia , Fígado Gorduroso/patologia , Fígado Gorduroso/prevenção & controle , Feminino , Células Espumosas/efeitos dos fármacos , Células Espumosas/enzimologia , Células Espumosas/patologia , Células Hep G2 , Humanos , Hidrocarbonetos Fluorados/toxicidade , Hipertrigliceridemia/induzido quimicamente , Hipertrigliceridemia/enzimologia , Hipertrigliceridemia/patologia , Hipertrigliceridemia/prevenção & controle , Fígado/efeitos dos fármacos , Fígado/metabolismo , Receptores X do Fígado , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores Nucleares Órfãos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/toxicidade
2.
Int J Cancer ; 136(4): 771-83, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24947959

RESUMO

Several MEK1/2 inhibitors have been in clinical trial evaluation for cancer treatment. Interferon-γ (IFN-γ) is a cytokine with multiple biological functions including antitumor activity. Expression of IFN-γ can be induced by liver X receptor (LXR), a ligand-activated transcription factor. However, it remains unknown if the anti-cancer action of MEK1/2 inhibitors is completed, at least in part, by activating IFN-γ expression. In this study, we determined that U0126, a MEK1/2 inhibitor, increased tumor-free and survival rates and decreased growth of inoculated Lewis lung carcinomas in wild type mice. However, the protective effects were substantially attenuated in IFN-γ deficient (IFN-γ-/-) mice. At cellular and molecular levels, MEK1/2 inhibitors increased IFN-γ protein and mRNA expression and activated natural IFN-γ promoter but not the IFN-γ promoters with mutations of the LXR responsive elements (LXREs). MEK1/2 inhibitors also enhanced formation of the LXRE-nuclear protein complexes by inducing LXR expression and nuclear translocation. Similarly, MEK1/2 siRNA inhibited phosphorylation of ERK1/2 by MEK1/2 while activated IFN-γ expression. In contrast, inhibition of LXR expression by siRNA blocked MEK1/2 inhibitors-induced IFN-γ expression. U0126 also inhibited chemicals-induced pulmonary carcinomas, which was associated with increased IFN-γ expression in the lung. Taken together, our study suggests that MEK1/2 inhibitors induce IFN-γ production in an LXR-dependent manner and the induction of IFN-γ expression can partially contribute to the anti-tumorigenic properties of U0126.


Assuntos
Antineoplásicos/farmacologia , Butadienos/farmacologia , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Interferon gama/genética , Nitrilas/farmacologia , Ativação Transcricional/efeitos dos fármacos , Transporte Ativo do Núcleo Celular , Animais , Carcinoma Pulmonar de Lewis/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Feminino , Expressão Gênica , Interferon gama/metabolismo , Receptores X do Fígado , Pulmão/metabolismo , Pulmão/patologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Camundongos Endogâmicos C57BL , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cell Metab ; 6(6): 446-57, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18054314

RESUMO

The Akt signaling pathway controls several cellular functions in the cardiovascular system; however, its role in atherogenesis is unknown. Here, we show that the genetic ablation of Akt1 on an apolipoprotein E knockout background (ApoE(-/-)Akt1(-/-)) increases aortic lesion expansion and promotes coronary atherosclerosis. Mechanistically, lesion formation is due to the enhanced expression of proinflammatory genes and endothelial cell and macrophage apoptosis. Bone marrow transfer experiments showing that macrophages from ApoE(-/-)Akt1(-/-) donors were not sufficient to worsen atherogenesis when transferred to ApoE(-/-) recipients suggest that lesion expansion in the ApoE(-/-)Akt1(-/-) strain might be of vascular origin. In the vessel wall, the loss of Akt1 increases inflammatory mediators and reduces eNOS phosphorylation, suggesting that Akt1 exerts vascular protection against atherogenesis. The presence of coronary lesions in ApoE(-/-)Akt1(-/-) mice provides a new model for studying the mechanisms of acute coronary syndrome in humans.


Assuntos
Aterosclerose/etiologia , Oclusão Coronária/etiologia , Proteínas Proto-Oncogênicas c-akt/deficiência , Síndrome Coronariana Aguda/etiologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Apolipoproteínas E/fisiologia , Apoptose , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Transplante de Medula Óssea , Oclusão Coronária/genética , Oclusão Coronária/patologia , Oclusão Coronária/fisiopatologia , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/patologia , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/fisiologia
4.
EMBO J ; 27(14): 1932-43, 2008 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-18566586

RESUMO

The target of rapamycin (TOR), as part of the rapamycin-sensitive TOR complex 1 (TORC1), regulates various aspects of protein synthesis. Whether TOR functions in this process as part of TORC2 remains to be elucidated. Here, we demonstrate that mTOR, SIN1 and rictor, components of mammalian (m)TORC2, are required for phosphorylation of Akt and conventional protein kinase C (PKC) at the turn motif (TM) site. This TORC2 function is growth factor independent and conserved from yeast to mammals. TM site phosphorylation facilitates carboxyl-terminal folding and stabilizes newly synthesized Akt and PKC by interacting with conserved basic residues in the kinase domain. Without TM site phosphorylation, Akt becomes protected by the molecular chaperone Hsp90 from ubiquitination-mediated proteasome degradation. Finally, we demonstrate that mTORC2 independently controls the Akt TM and HM sites in vivo and can directly phosphorylate both sites in vitro. Our studies uncover a novel function of the TOR pathway in regulating protein folding and stability, processes that are most likely linked to the functions of TOR in protein synthesis.


Assuntos
Proteína Quinase C/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Motivos de Aminoácidos , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Humanos , Camundongos , Modelos Moleculares , Complexos Multiproteicos/metabolismo , Dobramento de Proteína , Proteína Quinase C/química , Proteínas Proto-Oncogênicas c-akt/química , Serina-Treonina Quinases TOR
5.
Blood ; 116(24): 5423-33, 2010 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-20813898

RESUMO

Our previous work has shown that axon guidance gene family Nogo-B and its receptor (NgBR) are essential for chemotaxis and morphogenesis of endothelial cells in vitro. To investigate NogoB-NgBR function in vivo, we cloned the zebrafish ortholog of both genes and studied loss of function in vivo using morpholino antisense technology. Zebrafish ortholog of Nogo-B is expressed in somite while expression of zebrafish NgBR is localized in intersomitic vessel (ISV) and axial dorsal aorta during embryonic development. NgBR or Nogo-B knockdown embryos show defects in ISV sprouting in the zebrafish trunk. Mechanistically, we found that NgBR knockdown not only abolished its ligand Nogo-B-stimulated endothelial cell migration but also reduced the vascular endothelial growth factor (VEGF)-stimulated phosphorylation of Akt and vascular endothelial growth factor-induced chemotaxis and morphogenesis of human umbilical vein endothelial cells. Further, constitutively activated Akt (myristoylated [myr]Akt) or human NgBR can rescue the NgBR knockdown umbilical vein endothelial cell migration defects in vitro or NgBR morpholino-caused ISV defects in vivo. These data place Akt at the downstream of NgBR in both Nogo-B- and VEGF-coordinated sprouting of ISVs. In summary, this study identifies the in vivo functional role for Nogo-B and its receptor (NgBR) in angiogenesis in zebrafish.


Assuntos
Neovascularização Fisiológica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Superfície Celular/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Quimiotaxia , Desenvolvimento Embrionário , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Humanos , Fosforilação , RNA Antissenso/farmacologia , Receptores de Superfície Celular/genética , Fator A de Crescimento do Endotélio Vascular/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
6.
Blood ; 115(1): 133-9, 2010 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19880500

RESUMO

Recently, messenger RNAs in eukaryotes have shown to associate with antisense (AS) transcript partners that are often referred to as long noncoding RNAs (lncRNAs) whose function is largely unknown. Here, we have identified a natural AS transcript for tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-1 (tie-1), tie-1AS lncRNA in zebrafish, mouse, and humans. In embryonic zebrafish, tie-1AS lncRNA transcript is expressed temporally and spatially in vivo with its native target, the tie-1 coding transcript and in additional locations (ear and brain). The tie-1AS lncRNA selectively binds tie-1 mRNA in vivo and regulates tie-1 transcript levels, resulting in specific defects in endothelial cell contact junctions in vivo and in vitro. The ratio of tie-1 versus tie-1AS lncRNA is altered in human vascular anomaly samples. These results directly implicate noncoding RNA-mediated transcriptional regulation of gene expression as a fundamental control mechanism for physiologic processes, such as vascular development.


Assuntos
Loci Gênicos/genética , RNA Antissenso/genética , RNA não Traduzido/genética , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/genética , Animais , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio/efeitos dos fármacos , Endotélio/metabolismo , Endotélio/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Junções Intercelulares/efeitos dos fármacos , Junções Intercelulares/metabolismo , Junções Intercelulares/patologia , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos , Fenótipo , RNA Antissenso/metabolismo , RNA não Traduzido/metabolismo , Receptor de TIE-1/genética , Receptor de TIE-1/metabolismo , Especificidade da Espécie , Doenças Vasculares/genética , Doenças Vasculares/patologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteínas de Peixe-Zebra/genética
7.
Arterioscler Thromb Vasc Biol ; 28(1): 105-11, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17975117

RESUMO

OBJECTIVE: Heat-shock protein 90 (Hsp90) coordinates the regulation of diverse signaling proteins. We try to develop a new tool to explore the regulatory functions of Hsp90 in endothelial cells (ECs) instead of the existing chemical approaches. METHODS AND RESULTS: We designed a dominant-negative Hsp90 construct by site-direct mutagenesis of residue Asp-88 to Asn (D88N-Hsp90) based on the structure of the ATP/ADP-binding site. Recombinant wild-type Hsp90 protein binds ATP-Sepharose beads in manner inhibited by ATP or 17-AAG, a specific inhibitor for Hsp90, however the binding activity of D88N-Hsp90 was markedly reduced and the inhibitory effects of ATP or 17-AAG were negligible. The dimerization between endogenous Hsp90alpha and exogenous HA-Hsp90beta was confirmed by immunoprecipitation, however the association between eNOS and D88N-Hsp90 was less than WT-Hsp90. Furthermore, adenoviral transduction of bovine aortic ECs with D88N-Hsp90 suppressed VEGF-induced phosphorylation of Akt, eNOS, and NO release and the inhibitory effect was blocked by okadaic acid. Moreover, D88N-Hsp90 abolished VEGF-stimulated Rac activation and suppressed VEGF-induced stress fiber formation. Transduction with D88N-Hsp90 decreased growth medium mediated migration of wild-type ECs, but not Akt1(-/-) ECs suggesting that Akt is key target of Hsp90. CONCLUSIONS: Our data demonstrate that dominant-negative Hsp90 modulates endothelial cell mobility mainly through PP2A-mediated dephosphorylation of Akt and Rac activation.


Assuntos
Movimento Celular/fisiologia , Células Endoteliais/enzimologia , Proteínas de Choque Térmico HSP90/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia , Adenoviridae , Animais , Bovinos , Células Cultivadas , Pulmão/citologia , Camundongos , Mutagênese Sítio-Dirigida , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais
8.
Diabetes ; 68(9): 1841-1852, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31217174

RESUMO

Damage to the vasculature is the primary mechanism driving chronic diabetic microvascular complications such as diabetic nephropathy, which manifests as albuminuria. Therefore, treatments that protect the diabetic vasculature have significant therapeutic potential. Soluble neurite outgrowth inhibitor-B (sNogo-B) is a circulating N-terminus isoform of full-length Nogo-B, which plays a key role in vascular remodeling following injury. However, there is currently no information on the role of sNogo-B in the context of diabetic nephropathy. We demonstrate that overexpression of sNogo-B in the circulation ameliorates diabetic kidney disease by reducing albuminuria, hyperfiltration, and abnormal angiogenesis and protecting glomerular capillary structure. Systemic sNogo-B overexpression in diabetic mice also associates with dampening vascular endothelial growth factor-A signaling and reducing endothelial nitric oxide synthase, AKT, and GSK3ß phosphorylation. Furthermore, sNogo-B prevented the impairment of tube formation, which occurred when human endothelial cells were exposed to sera from patients with diabetic kidney disease. Collectively, these studies provide the first evidence that sNogo-B protects the vasculature in diabetes and may represent a novel therapeutic target for diabetic vascular complications.


Assuntos
Capilares/metabolismo , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Glomérulos Renais/irrigação sanguínea , Proteínas Nogo/metabolismo , Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Animais , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/genética , Humanos , Glomérulos Renais/metabolismo , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Nogo/sangue , Proteínas Nogo/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
Br J Pharmacol ; 175(9): 1486-1503, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29394501

RESUMO

BACKGROUND AND PURPOSE: The liver X receptor (LXR) agonist T317 reduces atherosclerosis but induces fatty liver. Metformin activates energy metabolism by activating AMPKα. In this study, we determined if interactions between metformin and T317 could inhibit atherosclerosis without activation of hepatic lipogenesis. EXPERIMENTAL APPROACH: Apolipoprotein E-deficient mice were treated with T317, metformin or both agents, in a high-fat diet for 16 weeks. Then, samples of aorta, liver, macrophage and serum were collected to determine atherosclerotic lesions, fatty liver, lipid profiles and expression of related proteins. Techniques used included immunohistochemistry, histology, qRT-PCR and Western blot. KEY RESULTS: T317 inhibited en face and aortic root sinus lesions, and the inhibition was further enhanced by addition of metformin. Co-treatment with metformin and T317 increased lesion stability, by increasing collagen content, and reducing necrotic cores and calcification. Formation of macrophages/foam cells and their accumulation in arterial wall were inhibited by the co-treatment, which was accompanied by increased ABCA1/ABCG1 expression, reduced monocyte adhesion and apparent local proliferation of macrophages. Metformin blocked T317-induced fatty liver by inhibiting T317-induced hepatic LXRα nuclear translocation and expression of lipogenic genes and by activating AMPKα. Moreover, co-treatment with T317 and metformin improved triglyceride metabolism by inducing expression of adipose triglyceride lipase, hormone-sensitive lipase, PPARα and carnitine acetyltransferase and by inhibiting acyl-CoA:diacylglycerol acyltransferase 1 expression. CONCLUSIONS AND IMPLICATIONS: Co-treatment with T317 and metformin inhibited the development of atherosclerosis without activation of lipogenesis, suggesting that combined treatment with T317 and metformin may be a novel approach to inhibition of atherosclerosis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Receptores X do Fígado/agonistas , Metformina/farmacologia , Transportador 1 de Cassete de Ligação de ATP/biossíntese , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/biossíntese , Animais , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/enzimologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Adesão Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Dieta Hiperlipídica , Interações Medicamentosas , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/prevenção & controle , Células Espumosas/efeitos dos fármacos , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Receptores X do Fígado/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Monócitos/efeitos dos fármacos
11.
Biol Open ; 4(1): 48-61, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25505152

RESUMO

In this study, we have identified a novel member of the AMPK family, namely Sucrose non-fermenting related kinase (Snrk), that is responsible for maintaining cardiac metabolism in mammals. SNRK is expressed in the heart, and brain, and in cell types such as endothelial cells, smooth muscle cells and cardiomyocytes (CMs). Snrk knockout (KO) mice display enlarged hearts, and die at postnatal day 0. Microarray analysis of embryonic day 17.5 Snrk hearts, and blood profile of neonates display defect in lipid metabolic pathways. SNRK knockdown CMs showed altered phospho-acetyl-coA carboxylase and phospho-AMPK levels similar to global and endothelial conditional KO mouse. Finally, adult cardiac conditional KO mouse displays severe cardiac functional defects and lethality. Our results suggest that Snrk is essential for maintaining cardiac metabolic homeostasis, and shows an autonomous role for SNRK during mammalian development.

12.
Blood ; 100(9): 3245-52, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12384424

RESUMO

Kallistatin is a unique serine proteinase inhibitor (serpin) and a heparin-binding protein. It has been localized in vascular smooth muscle cells and endothelial cells of human blood vessels, suggesting that kallistatin may be involved in the regulation of vascular function. Our previous study showed that kallistatin plays a role in neointima hyperplasia. In this study, we investigated the potential role of kallistatin in angiogenesis in vitro and in vivo. Purified human kallistatin significantly inhibited vascular endothelial growth factor (VEGF)- or basic fibroblast growth factor (bFGF)-induced proliferation, migration, and adhesion of cultured endothelial cells. Kallistatin attenuated VEGF- or bFGF-induced capillary density and hemoglobin content in subcutaneously implanted Matrigel plugs in mice. To further investigate the role of kallistatin in angiogenesis, we prepared adenovirus carrying the human kallistatin cDNA (Ad.HKBP) and evaluated the effect of kallistatin gene delivery on spontaneous angiogenesis in a rat model of hind-limb ischemia. Local kallistatin gene delivery significantly reduced capillary formation and regional blood perfusion recovery in the ischemic hind limb after removal of the femoral artery. Furthermore, a single intratumoral injection of Ad.HKBP into pre-established human breast tumor xenografts grown in athymic mice resulted in significant inhibition of tumor growth. CD31 immunostaining of tumor sections showed a decreased number of blood vessels in the kallistatin-treated group as compared to the control. These results demonstrate a novel role of kallistatin in the inhibition of angiogenesis and tumor growth.


Assuntos
Inibidores da Angiogênese/fisiologia , Proteínas de Transporte/fisiologia , Serpinas/fisiologia , Adenocarcinoma/patologia , Adenoviridae/genética , Inibidores da Angiogênese/genética , Animais , Neoplasias da Mama/patologia , Proteínas de Transporte/genética , Adesão Celular , Divisão Celular , Movimento Celular , Células Cultivadas , DNA Complementar/administração & dosagem , DNA Complementar/genética , Implantes de Medicamento , Fatores de Crescimento Endotelial/administração & dosagem , Fatores de Crescimento Endotelial/antagonistas & inibidores , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/citologia , Feminino , Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/farmacologia , Vetores Genéticos/administração & dosagem , Membro Posterior/irrigação sanguínea , Humanos , Injeções Intralesionais , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Isquemia/fisiopatologia , Linfocinas/administração & dosagem , Linfocinas/antagonistas & inibidores , Linfocinas/farmacologia , Masculino , Neovascularização Patológica/terapia , Neovascularização Fisiológica/efeitos dos fármacos , Ratos , Proteínas Recombinantes de Fusão/fisiologia , Serpinas/genética , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Am J Physiol Cell Physiol ; 284(6): C1604-13, 2003 06.
Artigo em Inglês | MEDLINE | ID: mdl-12734113

RESUMO

Kallistatin is a serpin first identified as a specific inhibitor of tissue kallikrein. Our recent studies showed that kallikrein promoted angiogenesis, whereas kallistatin inhibited angiogenesis and tumor growth. This study is aimed to identify the structural elements of kallistatin essential for its antiangiogenic function. Kallistatin mutants at the hinge region (A377T) and a major heparin-binding domain (K312A/K313A) were created by site-directed mutagenesis. Recombinant kallistatin mutant A377T did not bind or inhibit tissue kallikrein activity. Wild-type kallistatin and kallistatin mutant A377T, but not kallistatin mutant K312A/K313A lacking heparin-binding activity, inhibited VEGF-induced proliferation, growth, and migration of human microvascular endothelial cells. Similarly, wild-type kallistatin and kallistatin mutant A337T, but not kallistatin mutant K312A/K313A, significantly inhibited VEGF-induced capillary tube formation of cultured endothelial cells in Matrigel and capillary formation in Matrigel implants in mice. To elucidate the role of the heparin-binding domain in modulating angiogenesis, we showed that wild-type kallistatin interrupted the binding of (125)I-labeled VEGF to endothelial cells, whereas kallistatin mutant K312A/K313A did not interfere with VEGF binding. Consequently, wild-type kallistatin, but not kallistatin mutant K312A/K313A, suppressed VEGF-induced phosphorylation of Akt. Taken together, these results indicate that the heparin-binding domain, but not the reactive site loop of kallistatin, is essential for inhibiting VEGF-induced angiogenesis.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Neovascularização Fisiológica , Serpinas/química , Serpinas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/genética , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Células Cultivadas , Colágeno , Combinação de Medicamentos , Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Calicreínas/antagonistas & inibidores , Calicreínas/metabolismo , Laminina , Linfocinas/metabolismo , Substâncias Macromoleculares , Masculino , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Proteoglicanas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serpinas/genética , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA