Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nat Med ; 12(7): 793-800, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16799557

RESUMO

Vascular endothelial growth factor (VEGF) exerts crucial functions during pathological angiogenesis and normal physiology. We observed increased hematocrit (60-75%) after high-grade inhibition of VEGF by diverse methods, including adenoviral expression of soluble VEGF receptor (VEGFR) ectodomains, recombinant VEGF Trap protein and the VEGFR2-selective antibody DC101. Increased production of red blood cells (erythrocytosis) occurred in both mouse and primate models, and was associated with near-complete neutralization of VEGF corneal micropocket angiogenesis. High-grade inhibition of VEGF induced hepatic synthesis of erythropoietin (Epo, encoded by Epo) >40-fold through a HIF-1alpha-independent mechanism, in parallel with suppression of renal Epo mRNA. Studies using hepatocyte-specific deletion of the Vegfa gene and hepatocyte-endothelial cell cocultures indicated that blockade of VEGF induced hepatic Epo by interfering with homeostatic VEGFR2-dependent paracrine signaling involving interactions between hepatocytes and endothelial cells. These data indicate that VEGF is a previously unsuspected negative regulator of hepatic Epo synthesis and erythropoiesis and suggest that levels of Epo and erythrocytosis could represent noninvasive surrogate markers for stringent blockade of VEGF in vivo.


Assuntos
Eritropoetina/fisiologia , Fígado/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Hematócrito , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , Modelos Animais , Policitemia/fisiopatologia , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Vasos Retinianos/fisiologia
2.
Nat Med ; 9(1): 47-52, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12483208

RESUMO

Cytokines can initiate and perpetuate human diseases, and are among the best-validated of therapeutic targets. Cytokines can be blocked by the use of soluble receptors; however, the use of this approach for cytokines such as interleukin (IL)-1, IL-4, IL-6 and IL-13 that use multi-component receptor systems is limited because monomeric soluble receptors generally exhibit low affinity or function as agonists. We describe here a generally applicable method to create very high-affinity blockers called 'cytokine traps' consisting of fusions between the constant region of IgG and the extracellular domains of two distinct cytokine receptor components involved in binding the cytokine. Traps potently block cytokines in vitro and in vivo and represent a substantial advance in creating novel therapeutic candidates for cytokine-driven diseases.


Assuntos
Antígenos CD/metabolismo , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Fragmentos Fc das Imunoglobulinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Interleucina-6/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Divisão Celular/fisiologia , Linhagem Celular , Receptor gp130 de Citocina , Citocinas/imunologia , Dimerização , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Macaca fascicularis , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos , Ligação Proteica , Distribuição Aleatória , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo
3.
Proc Natl Acad Sci U S A ; 104(47): 18363-70, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18000042

RESUMO

VEGF is the best characterized mediator of tumor angiogenesis. Anti-VEGF agents have recently demonstrated impressive efficacy in human cancer trials, but the optimal dosing of such agents must still be determined empirically, because biomarkers to guide dosing have yet to be established. The widely accepted (but unverified) assumption that VEGF production is quite low in normal adults led to the notion that increased systemic VEGF levels might quantitatively reflect tumor mass and angiogenic activity. We describe an approach to determine host and tumor production of VEGF, using a high-affinity and long-lived VEGF antagonist now in clinical trials, the VEGF Trap. Unlike antibody complexes that are usually rapidly cleared, the VEGF Trap forms inert complexes with tissue- and tumor-derived VEGF that remain stably in the systemic circulation, where they are readily assayable, providing unprecedented capability to accurately measure VEGF production. We report that VEGF production is surprisingly high in non-tumor-bearing rodents and humans, challenging the notion that systemic VEGF levels can serve as a sensitive surrogate for tumor load; tumor VEGF contribution becomes significant only with very large tumor loads. These findings have the important corollary that anti-VEGF therapies must be sufficiently dosed to avoid diversion by host-derived VEGF. We further show that our assay can indicate when VEGF is optimally blocked; such biomarkers to guide dosing do not exist for other anti-VEGF agents. Based on this assay, VEGF Trap doses currently being assessed in clinical trials are in the efficacious range.


Assuntos
Inibidores da Angiogênese/farmacologia , Fatores de Crescimento do Endotélio Vascular/biossíntese , Envelhecimento/fisiologia , Inibidores da Angiogênese/imunologia , Animais , Anticorpos/imunologia , Biomarcadores , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos SCID , Ligação Proteica , Fatores de Crescimento do Endotélio Vascular/sangue , Fatores de Crescimento do Endotélio Vascular/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto , Displasia do Colo do Útero/metabolismo , Displasia do Colo do Útero/patologia
4.
Kidney Int ; 74(3): 300-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18480750

RESUMO

The loss of interstitial capillaries is a feature of several experimental models of renal disease and this contributes to secondary kidney injury. Angiopoietin-1 is a secreted growth factor which binds to Tie-2 present on endothelia to enhance cell survival thereby stabilizing capillary architecture in-vitro. Previous studies showed that angiopoietin-1 prevented renal capillary and interstitial lesions following experimental ureteric obstruction. We tested here the effect of angiopoietin-1 treatment on capillary loss and associated tubulointerstitial damage known to follow recovery from folic acid-induced tubular necrosis and acute renal injury. We found that delivery of angiopoietin-1 by adenoviral vectors stabilized peritubular capillaries in folic acid nephropathy but this was accompanied by profibrotic and inflammatory effects. These results suggest that the use of endothelial growth factor therapy for kidney disease may have varying outcomes that depend on the disease model tested.


Assuntos
Angiopoietina-1/efeitos adversos , Fibrose/induzido quimicamente , Inflamação/induzido quimicamente , Necrose Tubular Aguda/tratamento farmacológico , Adenoviridae/genética , Angiopoietina-1/administração & dosagem , Angiopoietina-1/uso terapêutico , Animais , Modelos Animais de Doenças , Ácido Fólico/efeitos adversos , Vetores Genéticos , Nefropatias/induzido quimicamente , Nefropatias/tratamento farmacológico , Nefropatias/patologia , Necrose Tubular Aguda/induzido quimicamente , Necrose Tubular Aguda/patologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Circulação Renal/efeitos dos fármacos
5.
Sci Rep ; 8(1): 505, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29323190

RESUMO

Angiopoietin-1 (Ang1) and Angiopoietin-2 (Ang2) are ligands for Tie2, an endothelial-specific receptor tyrosine kinase that is an essential regulator of angiogenesis. Here we report the identification, via expression cloning, of thrombomodulin (TM) as another receptor for Ang1 and Ang2. Thrombomodulin is an endothelial cell surface molecule that plays an essential role as a coagulation inhibitor via its function as a cofactor in the thrombin-mediated activation of protein C, an anticoagulant protein, as well as thrombin-activatable fibrinolysis inhibitor (TAFI). Ang1 and Ang2 inhibited the thrombin/TM-mediated generation of activated protein C and TAFI in cultured endothelial cells, and inhibited the binding of thrombin to TM in vitro. Ang2 appears to bind TM with higher affinity than Ang1 and is a more potent inhibitor of TM function. Consistent with a potential role for angiopoietins in coagulation, administration of thrombin to mice rapidly increased plasma Ang1 levels, presumably reflecting release from activated platelets (previously shown to contain high levels of Ang1). In addition, Ang1 levels were significantly elevated in plasma prepared from wound blood, suggesting that Ang1 is released from activated platelets at sites of vessel injury. Our results imply a previously undescribed role for angiopoietins in the regulation of hemostasis.


Assuntos
Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Trombina/metabolismo , Trombomodulina/metabolismo , Angiopoietina-1/sangue , Angiopoietina-1/genética , Angiopoietina-2/genética , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Células COS , Carboxipeptidase B2/metabolismo , Chlorocebus aethiops , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fator Plaquetário 4/metabolismo , Ligação Proteica , Proteína C/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptor TIE-2/antagonistas & inibidores , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Trombina/química , Trombina/farmacologia , Trombomodulina/genética
6.
Circulation ; 110(16): 2430-5, 2004 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-15477421

RESUMO

BACKGROUND: The rate of reendothelialization is critical in neointima formation after arterial injury. Vascular endothelial growth factor (VEGF), a potent endothelial mitogen, has been advocated for accelerating endothelial repair and preventing intimal hyperplasia after percutaneous coronary interventions. However, the precise mechanism of action of VEGF treatment and the physiologic role of endogenous VEGF after arterial injury are not well described. To better understand the role of VEGF in arterial repair, we overexpressed both VEGF and a soluble, chimeric VEGF receptor (VEGF-trap), which binds free VEGF with high affinity, in a mouse model of arterial injury. METHODS AND RESULTS: Four groups of C57BL/6 mice underwent denuding endothelial injury 1 day after systemic injection of recombinant adenovirus expressing (1) VEGF, (2) VEGF-trap, (3) VEGF plus VEGF-trap, or (4) control adenovirus. Circulating levels of adenovirus-encoded proteins were significantly elevated after gene transfer. VEGF overexpression accelerated reendothelialization and increased luminal endothelial cell proliferation 2 weeks after arterial injury (P<0.05), resulting in decreased neointima formation at 4 weeks compared with control (P<0.01). Cotreatment with VEGF-trap completely sequestered free VEGF and abrogated the beneficial effect of VEGF overexpression. Interestingly, sequestration of endogenous VEGF by VEGF-trap overexpression alone also led to delayed reendothelialization at 2 weeks (P<0.01) and increased neointima formation at 4 weeks (P<0.01). CONCLUSIONS: VEGF overexpression accelerated endothelial repair and inhibited neointima formation after arterial injury. Conversely, sequestration of exogenous and/or endogenous VEGF by VEGF-trap delayed reendothelialization and significantly increased neointima size. This demonstrates the therapeutic potential of VEGF but also emphasizes the important physiologic role of endogenous VEGF in vascular repair.


Assuntos
Endotélio Vascular/lesões , Terapia Genética , Fator A de Crescimento do Endotélio Vascular/fisiologia , Cicatrização/fisiologia , Angioplastia/efeitos adversos , Animais , Divisão Celular , Células Endoteliais/patologia , Endotélio Vascular/patologia , Humanos , Hiperplasia , Processamento de Imagem Assistida por Computador , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Método Simples-Cego , Túnica Íntima/patologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética
7.
J Clin Endocrinol Metab ; 90(2): 1114-22, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15562010

RESUMO

Follicular development is associated with intense angiogenesis and increased permeability of blood vessels under the control of locally produced angiogenic factors such as vascular endothelial growth factor (VEGF). The aim of the present study was to evaluate the effects of transient inhibition of VEGF on pituitary-ovarian function in the macaque. Animals were given a single, iv injection of a potent, receptor-based VEGF antagonist, the VEGF Trap. VEGF Trap was given at a dose of 4, 1, or 0.25 mg/kg in the midfollicular phase or at 1.0 mg/kg in the late follicular phase. Controls were treated with vehicle or a control protein, recombinant human Fc (1 mg/kg). Blood samples were collected once daily for 12 d after injection, and three times per week thereafter until normal ovulatory cycles had resumed. The VEGF Trap produced a rapid suppression of estradiol and inhibin B concentrations at all doses tested, followed by a marked and sustained increase in LH and FSH. Ovulation and formation of a functional corpus luteum, as evidenced by increased serum progesterone levels, failed to occur at the anticipated time. Normal ovarian activity resumed when plasma concentrations of unbound VEGF Trap fell below about 1 mg/liter. When treatment was initiated in the midfollicular phase, control macaques ovulated 7.2 +/- 0.4 d later, but ovulation was delayed in a dose-dependent manner by VEGF Trap, occurring 23 +/- 0.7, 30 +/- 1.4, and 43 +/- 0.8 d after injection of 0.25, 1, or 4 mg/kg, respectively. Thus, the VEGF Trap exerts a potent, dose-dependent, but reversible inhibitory effect on ovarian function.


Assuntos
Ovário/fisiologia , Ovulação/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Estradiol/fisiologia , Antagonistas de Estrogênios/farmacologia , Feminino , Inibinas/antagonistas & inibidores , Injeções Intravenosas , Macaca , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Ovário/efeitos dos fármacos , Receptores de Fatores de Crescimento , Proteínas Recombinantes de Fusão/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/administração & dosagem
8.
J Am Coll Cardiol ; 44(4): 897-903, 2004 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-15312878

RESUMO

OBJECTIVES: The aim of this research was to test the effects of vascular endothelial growth factor (VEGF)/angiopoietin-1 (Ang-1) on adult hypoperfused tissues. BACKGROUND: Angiopoietin-1 and VEGF act separately and synergistically in vascular development during embryogenesis. However, little is known regarding their relative roles in collateral development after chronic arterial obstruction and tissue ischemia in the adult. METHODS: Central and caudal ear arteries of 32 rabbits were ligated to induce ischemia. At two months, when flow was about 65% of pre-ligation values, we injected intradermally 10(9) plaque-forming unit adenovirus with the following transgenes: Ang-1, VEGF, or a combination of both. Ear perfusion was followed up for four weeks, and vessel leakage was assessed by Evens Blue test. RESULTS: Before injection, flow was 65% of baseline, and endogenous VEGF levels in ischemic tissue were increased. Adenovirus-encoding VEGF gene (Ad.VEGF) at one week caused a visible inflammatory response associated with a 24% flow increase (p = 0.018). Adenovirus-encoding Ang-1 gene (Ad.Ang-1) increased flow 22% (p = 0.004) with no visible inflammation; Ad.VEGF caused three times as much vessel leakage as Ad.Ang-1 (142.5 +/- 38 vs. 49.5 +/- 9.8 ng Evens Blue/mg tissue; p < 0.001). However, at four weeks, compared with baseline, VEGF decreased flow 18% (p = 0.004), whereas Ang-1 increased tissue perfusion 26% (p < 0.001). This effect was abolished when Ad.Ang-1 was injected with soluble VEGF receptor [Ad.Flt(1-3)-Fc], which blocks VEGF-dependent signaling. Exogenous Ang-1 did not increase perfusion in a normally perfused ear, in which endogenous VEGF is not expressed. CONCLUSIONS: Exogenous Ang-1 enhances perfusion in hypoperfused tissues only in the presence of increased levels of endogenous VEGF. Overexpression of VEGF, however, after causing an inflammatory response, does not improve collateral blood flow.


Assuntos
Indutores da Angiogênese/farmacologia , Angiopoietina-1/farmacologia , Endotélio Vascular/efeitos dos fármacos , Isquemia/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Adenoviridae/genética , Animais , Orelha Externa/irrigação sanguínea , Expressão Gênica , Terapia Genética , Isquemia/fisiopatologia , Masculino , Neovascularização Fisiológica/genética , Coelhos , Distribuição Aleatória , Fluxo Sanguíneo Regional
9.
Endocrinology ; 143(7): 2797-807, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12072415

RESUMO

This study was designed to investigate the effects of inhibition of thecal angiogenesis on follicular development in the marmoset monkey (Callithrix jacchus). To inhibit vascular endothelial growth factor (VEGF), a soluble combined truncated form of the fms-like tyrosine kinase (Flt) and kinase insert domain-containing receptor (KDR) receptor fused to IgG (VEGF Trap R1R2) was administered for 10 d during the follicular phase of the cycle. Changes in angiogenesis and follicular cell proliferation were quantified using immunocytochemistry for bromodeoxyuridine to obtain a proliferation index, CD31 to visualize endothelial cell area, and dual staining to distinguish thecal endothelial cell proliferation. The effects of the treatment on follicular development were assessed by morphometric analyses by measuring follicle diameter, thecal thickness, and a proliferation index for granulosa cells. Follicular atresia was detected and quantified using the terminal deoxynucleotidyltransferase-UTP nick end labeling method. Effects on gene expression of VEGF and its receptors, Flt and KDR, were studied by in situ hybridization. VEGF Trap R1R2 treatment resulted in a significant decrease in thecal proliferation and endothelial cell area, demonstrating the suppression of thecal angiogenesis. The absence of a normal thecal vasculature was associated with a significantly reduced thecal thickness. Antral follicular development was severely compromised, as indicated by decreased granulosa cell proliferation, decreased follicular diameter, and lack of development of ovulatory follicles. Furthermore, the rate of atresia was significantly increased. VEGF expression in granulosa and thecal cells increased after treatment, whereas Flt and KDR expressions in thecal endothelial cells were markedly decreased. These results show that VEGF Trap treatment is associated with the suppression of follicular angiogenesis, which results in the inhibition of antral follicular development and ovulation.


Assuntos
Inibidores da Angiogênese , Neovascularização Patológica/prevenção & controle , Folículo Ovariano/fisiologia , Ovulação/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Tumor da Célula Tecal/irrigação sanguínea , Animais , Apoptose/efeitos dos fármacos , Callithrix , Fatores de Crescimento Endotelial/metabolismo , Feminino , Células da Granulosa/efeitos dos fármacos , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Linfocinas/metabolismo , Neovascularização Patológica/patologia , Folículo Ovariano/efeitos dos fármacos , Receptores de Fatores de Crescimento , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
10.
Eur J Neurosci ; 4(6): 459-471, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-12106332

RESUMO

Cultured astrocytes are known to possess a range of neurotrophic activities in culture. In order to examine which factors may be responsible for these activities, we have examined the expression of the genes for four known neurotrophic factors-ciliary neurotrophic factor (CNTF), nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF) and neurotrophin 3 (NT3)-in purified astrocyte cultures derived from neonatal rat hippocampus. Hippocampal astrocytes were found to express mRNA for three neurotrophic factors-CNTF, NGF and NT3-at significantly higher levels than other cultured cell types or cell lines examined. BDNF messenger RNA (mRNA), however, was undetectable in these astrocytes. The levels of CNTF, NGF and NT3 mRNA in astrocytes were largely unaffected by their degree of confluency, while serum removal caused only a transient decrease in mRNA levels, which returned to basal levels within 48 h. Astrocyte-derived CNTF was found to comigrate with recombinant rat CNTF at 23 kD on a Western blot. Immunocytochemical analysis revealed strong CNTF immunoreactivity in the cytoplasm of astrocytes, weak staining in the nucleus, but no CNTF at the cell surface. NGF and NT3 were undetectable immunocytochemically. CNTF-like activity, as assessed by bioassay on ciliary ganglion neurons, was found in the extract of cultured astrocytes but not in conditioned medium, whereas astrocyte-conditioned medium supported survival of dorsal root ganglion neurons but not ciliary or nodose ganglion neurons. This conditioned medium activity was neutralized with antibodies to NGF. Astrocyte extract also supported survival of dorsal root ganglion and nodose ganglion neurons, but these activities were not blocked by anti-NGF. Part, but not all, of the activity in astrocyte extracts which sustained nodose ganglion neurons could be attributed to CNTF.

11.
Mol Cancer Ther ; 13(5): 1345-55, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24634416

RESUMO

EGFR blocking antibodies are approved for the treatment of colorectal cancer and head and neck squamous cell carcinoma (HNSCC). Although ERBB3 signaling has been proposed to limit the effectiveness of EGFR inhibitors, the underlying molecular mechanisms are not fully understood. To gain insight into these mechanisms, we generated potent blocking antibodies against ERBB3 (REGN1400) and EGFR (REGN955). We show that EGFR and ERBB3 are coactivated in multiple HNSCC cell lines and that combined blockade of EGFR and ERBB3 inhibits growth of these cell lines more effectively than blockade of either receptor alone. Blockade of EGFR with REGN955 strongly inhibited activation of ERK in HNSCC cell lines, whereas blockade of ERBB3 with REGN1400 strongly inhibited activation of Akt; only the combination of the 2 antibodies blocked both of these essential downstream pathways. We used a HER2 blocking antibody to show that ERBB3 phosphorylation in HNSCC and colorectal cancer cells is strictly dependent on association with HER2, but not EGFR, and that neuregulin 1 activates ERBB3/HER2 signaling to reverse the effect of EGFR blockade on colorectal cancer cell growth. Finally, although REGN1400 and REGN955 as single agents slowed the growth of HNSCC and colorectal cancer xenografts, the combination of REGN1400 plus REGN955 caused significant tumor regression. Our results indicate that activation of the Akt survival pathway by ERBB3/HER2 limits the effectiveness of EGFR inhibition, suggesting that REGN1400, which is currently in a phase I clinical trial, could provide benefit when combined with EGFR blocking antibodies.


Assuntos
Neoplasias Colorretais/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Camundongos , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-3/antagonistas & inibidores , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Endocrinology ; 153(4): 1972-83, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22334711

RESUMO

Using specific inhibitors established that angiogenesis in the ovarian follicle and corpus luteum is driven by vascular endothelial growth factor. Recently, it has been demonstrated that the Notch ligand, delta-like ligand 4 (Dll4) negatively regulates vascular endothelial growth factor-mediated vessel sprouting and branching. To investigate the role of Dll4 in regulation of the ovarian vasculature, we administered a neutralizing antibody to Dll4 to marmosets at the periovulatory period. The vasculature was examined on luteal d 3 or d 10: angiogenesis was determined by incorporation of bromodeoxyuridine, staining for CD31 and cell death by staining for activated caspase-3. Ovulatory progesterone rises were monitored to determine effects of treatment on luteal function and time to recover normal cycles in a separate group of animals. Additionally, animals were treated in the follicular or midluteal phase to determine effects of Dll4 inhibition on follicular development and luteal function. Controls were treated with human IgG (Fc). Corpora lutea from marmosets treated during the periovulatory period exhibited increased angiogenesis and increased vascular density on luteal d 3, but plasma progesterone was significantly suppressed. By luteal d 10, corpora lutea in treated ovaries were significantly reduced in size, with involution of luteal cells, increased cell death, and suppressed plasma progesterone concentrations. In contrast, initiation of anti-Dll4 treatment during the midluteal phase produced only a slight suppression of progesterone for the remainder of the cycle. Moreover, Dll4 inhibition had no appreciable effect on follicular development. These results show that Dll4 has a specific and critical role in the development of the normal luteal vasculature.


Assuntos
Callithrix/fisiologia , Corpo Lúteo/irrigação sanguínea , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Luteólise/fisiologia , Proteínas de Membrana/antagonistas & inibidores , Neovascularização Fisiológica/fisiologia , Ovário/fisiologia , Animais , Anticorpos Neutralizantes/farmacologia , Apoptose/fisiologia , Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/patologia , Feminino , Imunoglobulina G/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/fisiologia , Modelos Animais , Neovascularização Fisiológica/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Progesterona/sangue , Receptores Notch/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia
14.
J Vasc Res ; 44(4): 283-91, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17406120

RESUMO

BACKGROUND: Infusion of exogenous vascular endothelial growth factor (VEGF) into adult brain at doses above 60 ng/day induces dramatic angiogenesis accompanied by vascular leak and inflammation. Blood vessels formed by this treatment are dilated and tortuous, exhibiting a pathological morphology. Pathological VEGF-induced angiogenesis is preceded by vascular leak and inflammation, which have been proposed to mediate subsequent angiogenesis. METHODS: To test this hypothesis, we infused VEGF into the brains of adult rats to induce pathological angiogenesis. Some of these rats were treated with dexamethasone, a potent anti-inflammatory glucocorticoid, to inhibit inflammation and edema. RESULTS: We demonstrate that inhibition of inflammation by treatment with dexamethasone significantly attenuated VEGF-induced pathological angiogenesis. To present converging evidence that inflammation may be important in this angiogenic process, we also demonstrate that mice genetically deficient in the inflammatory mediator intercellular adhesion molecule-1 have attenuated VEGF-induced angiogenesis. These same mice showed normal amounts of physiological angiogenesis in response to enriched environments, however, suggesting that a generalized reduction in vascular plasticity could not account for their poor angiogenic response to VEGF. CONCLUSIONS: Taken together, the data from these experiments suggest that the inflammation which occurs before or during VEGF-induced pathological brain angiogenesis plays a contributory role in the pathological angiogenic process.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Neovascularização Patológica/tratamento farmacológico , Fatores Etários , Animais , Encéfalo/irrigação sanguínea , Encéfalo/imunologia , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neovascularização Patológica/induzido quimicamente , Neovascularização Patológica/imunologia , Ratos , Ratos Sprague-Dawley , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
15.
Microcirculation ; 13(6): 499-509, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16864416

RESUMO

OBJECTIVE: To determine which elements of the angiogenic process are controlled by VEGF under physiological conditions. METHODS: VEGF Trap was administered at 10 mg x kg(-1) by subcutaneous injection twice weekly to mice, which were subject to one of two established angiogenic stimuli: (1) increasing blood flow by administration of prazosin (50 mg L(-1)); (2) muscle overload by extirpation of a synergist. Angiogenesis was determined by calculating capillary to fiber ratio (C:F), and proliferating cell nuclear antigen (PCNA) staining localized to capillaries or the interstitium used to measure cell proliferation. Characteristic ultrastructural changes were quantified using electron microscopy. RESULTS: Administration of VEGF Trap abolished the increases in C:F seen in both models, and prevented growth of luminal filopodia and large vacuole formation. Overload-induced proliferation associated with capillaries was reduced, along with endothelial cell number abluminal sprouts and basement membrane breakage. However, interstitial proliferation remained high, along with the increased capillary association of pericytes and fibroblasts. CONCLUSIONS: VEGF is required for both models of angiogenesis, although some morphological changes lie upstream, or are independent of, VEGF involvement. The abolition of angiogenesis in a model unaffected by NO inhibition shows that NO is not required for all VEGF-dependent angiogenesis in vivo.


Assuntos
Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Fibroblastos/metabolismo , Masculino , Camundongos , Músculo Esquelético/metabolismo , Óxido Nítrico/metabolismo , Pericitos/metabolismo , Prazosina/farmacologia , Receptores de Fatores de Crescimento , Estresse Mecânico
16.
Reproduction ; 132(4): 589-600, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17008470

RESUMO

The intense angiogenesis characteristic of early corpus luteum development is dependent upon vascular endothelial growth factor (VEGF) as inhibitors of VEGF administered at the peri-ovulatory period suppress endothelial cell proliferation and progesterone secretion. We now report that administration of VEGF Trap, a soluble decoy receptor-based inhibitor, at the mid- or the late luteal phase in the marmoset results in a rapid decline in plasma progesterone. Since vascularisation of the corpus luteum is largely complete by the mid-luteal phase, it suggested that this functional luteolysis involved mechanisms other than inhibition of angiogenesis. A second experiment investigated the role of VEGF in maintaining the integrity of the luteal vasculature and hormone-producing cells. VEGF Trap was administered to marmosets in the mid-luteal phase and ovaries were obtained 1, 2, 4 or 8 days later for localisation of activated caspase-3 staining in the corpus luteum and compared with those obtained 2, 4 and 8 days after administration of control protein. The number of cells with activated caspase-3 staining was significantly increased after administration of VEGF Trap. Dual staining of activated caspase-3 with the endothelial cell marker CD31 showed that at 1 day post-treatment, more than 90% caspase-3-stained cells were vascular endothelium, prior to detection of an increasing incidence in death of hormone-producing cells on days 2 and 4. Staining with CD31 showed that the endothelial cell area was decreased after treatment. By 8 days after treatment, corpora lutea had regressed to varying degrees, while all control corpora lutea remained healthy. These results show that VEGF inhibition in the mid- or the late luteal phase induces functional luteolysis in the marmoset that is associated with premature and selective death of endothelial cells.


Assuntos
Inibidores da Angiogênese/farmacologia , Corpo Lúteo/irrigação sanguínea , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Animais , Callithrix , Morte Celular , Corpo Lúteo/citologia , Corpo Lúteo/efeitos dos fármacos , Células Endoteliais/patologia , Feminino , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica/métodos , Fase Luteal , Luteólise , Modelos Animais , Progesterona/sangue , Receptores de Fatores de Crescimento , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Proc Natl Acad Sci U S A ; 103(42): 15491-6, 2006 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-17030814

RESUMO

Angiopoietin (Ang)-2, a context-dependent agonist/antagonist for the vascular-specific Tie2 receptor, is highly expressed by endothelial cells at sites of normal and pathologic angiogenesis. One prevailing model suggests that in these settings, Ang-2 acts as an autocrine Tie2 blocker, inhibiting the stabilizing influence of the Tie2 activator Ang-1, thereby promoting vascular remodeling. However, the effects of endogenous Ang-2 on cells that are actively producing it have not been studied in detail. Here, we demonstrate that Ang-2 expression is rapidly induced in endothelial cells by the transcription factor FOXO1 after inhibition of the phosphatidylinositol 3-kinase/Akt pathway. We employ RNAi and blocking antibodies to show that in this setting, Ang-2 unexpectedly functions as a Tie2 agonist, bolstering Akt activity so as to provide negative feedback on FOXO1-regulated transcription and apoptosis. In addition, we show that Ang-2, like Ang-1, activates Tie2/Akt signaling in vivo, thereby inhibiting the expression of FOXO1 target genes. Consistent with a role for Ang-2 as a Tie2 activator, we demonstrate that Ang-2 inhibits vascular leak. Our data suggests a model in which Ang-2 expression is induced in stressed endothelial cells, where it acts as an autocrine Tie2 agonist and protective factor.


Assuntos
Angiopoietina-2/metabolismo , Comunicação Autócrina , Células Endoteliais/fisiologia , Estresse Oxidativo , Androstadienos/metabolismo , Angiopoietina-2/genética , Animais , Apoptose/fisiologia , Células Cultivadas , Células Endoteliais/citologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia , Ativação Transcricional , Wortmanina
18.
Blood ; 102(1): 161-8, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12649136

RESUMO

Gene therapy approaches involving vascular endothelial growth factor (VEGF) to promote therapeutic angiogenesis are under consideration for conditions ranging from ischemic heart disease to nonhealing skin ulcers. Here we make the surprising observation that the transgenic delivery of VEGF to the skin results in a profound inflammatory skin condition with many of the cellular and molecular features of psoriasis, including the characteristic vascular changes, epidermal alterations, and inflammatory infiltrates. Even longstanding psoriatic disease remains dependent on the transgenic VEGF in this model because it can be effectively reversed by the addition of VEGF Trap, a potent VEGF antagonist. Previous attempts to faithfully replicate the psoriatic phenotype through the transgenic delivery of epidermal keratinocyte growth factors or inflammatory mediators generated phenotypes with only partial resemblance to human psoriasis, leaving unanswered questions about the etiology of this disease. The ability of transgenic VEGF to induce a psoriasiform phenotype suggests a new etiology and treatment approach for this disease and further substantiates emerging concerns about possible proinflammatory adverse effects that might be associated with therapeutic attempts to deliver VEGF.


Assuntos
Fatores de Crescimento Endotelial/administração & dosagem , Fatores de Crescimento Endotelial/efeitos adversos , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/efeitos adversos , Linfocinas/administração & dosagem , Linfocinas/efeitos adversos , Psoríase/induzido quimicamente , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Modelos Animais de Doenças , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Humanos , Imuno-Histoquímica , Inflamação/induzido quimicamente , Inflamação/etiologia , Camundongos , Camundongos Transgênicos , Psoríase/etiologia , Receptores de Fatores de Crescimento , Proteínas Recombinantes de Fusão/farmacologia , Pele/efeitos dos fármacos , Pele/lesões , Pele/patologia , Transgenes , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
19.
J Cell Physiol ; 195(2): 241-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12652651

RESUMO

Vascular endothelial growth factor (VEGF) plays a central role in the development of retinal neovascularization and diabetic macular edema. There is also evidence suggesting that VEGF is an important stimulator for choroidal neovascularization. In this study, we investigated the effect of a specific inhibitor of VEGF, VEGF-TRAP(R1R2), in models for these disease processes. VEGF-TRAP(R1R2) is a fusion protein, which combines ligand binding elements taken from the extracellular domains of VEGF receptors 1 and 2 fused to the Fc portion of IgG1. Subcutaneous injections or a single intravitreous injection of VEGF-TRAP(R1R2) strongly suppressed choroidal neovascularization in mice with laser-induced rupture of Bruch's membrane. Subcutaneous injection of VEGF-TRAP(R1R2) also significantly inhibited subretinal neovascularization in transgenic mice that express VEGF in photoreceptors. In two models of VEGF-induced breakdown of the blood-retinal barrier (BRB), one in which recombinant VEGF is injected into the vitreous cavity and one in which VEGF expression is induced in the retina in transgenic mice, VEGF-TRAP(R1R2) significantly reduced breakdown of the BRB. These data confirm that VEGF is a critical stimulus for the development of choroidal neovascularization and indicate that VEGF-TRAP(R1R2) may provide a new agent for consideration for treatment of patients with choroidal neovascularization and diabetic macular edema.


Assuntos
Doenças da Coroide/tratamento farmacológico , Retinopatia Diabética/tratamento farmacológico , Fatores de Crescimento Endotelial/antagonistas & inibidores , Linfocinas/antagonistas & inibidores , Neovascularização Patológica/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Animais , Barreira Hematorretiniana/efeitos dos fármacos , Barreira Hematorretiniana/fisiologia , Corioide/efeitos dos fármacos , Corioide/metabolismo , Corioide/fisiopatologia , Doenças da Coroide/metabolismo , Doenças da Coroide/fisiopatologia , Retinopatia Diabética/metabolismo , Retinopatia Diabética/fisiopatologia , Modelos Animais de Doenças , Fatores de Crescimento Endotelial/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Linfocinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Receptores de Fatores de Crescimento/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Retina/efeitos dos fármacos , Retina/metabolismo , Retina/fisiopatologia , Artéria Retiniana/efeitos dos fármacos , Artéria Retiniana/patologia , Artéria Retiniana/fisiopatologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
20.
Genes Dev ; 18(9): 1060-71, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15132996

RESUMO

Despite genetic evidence establishing angiopoietin-1 (Ang-1) as an essential regulator of vascular development, the molecular mechanisms underlying Ang-1 function are almost completely uncharacterized. In this report, we demonstrate that Ang-1, via Akt activation, is a potent inhibitor of the forkhead transcription factor FKHR (FOXO1), identifying for the first time a nuclear signaling pathway through which Ang-1 modulates gene expression. We use microarray analysis to show that FKHR, whose function in endothelial cells has not previously been elucidated, regulates many genes associated with vascular destabilization and remodeling (including angiopoietin-2, an Ang-1 antagonist) and endothelial cell apoptosis (e.g., survivin, TRAIL). Ang-1 inhibits FKHR-mediated changes in gene expression and FKHR-induced apoptosis. Analysis of gene expression changes induced by an activated version of Akt confirms that FKHR is a major target through which Akt regulates transcription in endothelial cells. We use RNA interference to demonstrate that FKHR is required for the expression of genes (including Ang-2) that have important vascular functions. Our data suggest a novel, tissue-specific role for the Akt/FKHR pathway in the vasculature and suggest a mechanistic basis for the previously described actions of Ang-1 as a regulator of endothelial cell survival and blood vessel stability.


Assuntos
Angiopoietina-1/farmacologia , Proteínas de Ligação a DNA/fisiologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Proteínas Serina-Treonina Quinases , Fatores de Transcrição/fisiologia , Angiopoietina-1/fisiologia , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Bovinos , Células Cultivadas , DNA Complementar/genética , Proteínas de Ligação a DNA/genética , Endotélio Vascular/citologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Humanos , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA