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.
Am J Physiol Lung Cell Mol Physiol ; 306(1): L58-68, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24213918

RESUMO

Acute lung injury is marked by profound influx of activated neutrophils, which have delayed apoptosis, along with fluid accumulation that impairs lung function and causes high mortality. Inflammatory and antimicrobial molecules, such as reactive oxygen species from activated neutrophils with prolonged lifespan, cause tissue damage and contribute to lung dysfunction. Angiostatin, an endogenous antiangiogenic molecule, is expressed in the lavage fluid of patients with acute respiratory distress syndrome and modifies neutrophil infiltration in a mouse model of peritonitis. Our aim was to investigate the therapeutic role of angiostatin in acute lung injury. We analyzed bronchoalveolar lavage and lung tissues from C57BL/6 mouse model of Escherichia coli LPS-induced acute lung injury to assess the effects of angiostatin treatment. Subcutaneous angiostatin administered at 5 h after LPS treatment reduces histological signs of inflammation, protein accumulation, lung Gr1+ neutrophils, myeloperoxidase activity, and expression of phosphorylated p38 MAPK in lung tissues and peripheral blood neutrophils, while increasing the number of apoptotic cells in the lungs without affecting the levels of macrophage inflammatory protein-1 α, IL-1ß, keratinocyte chemoattractant, and monocyte chemoattractant protein-1 in lavage and lung homogenates at 9 and 24 h after LPS treatment. In contrast, angiostatin administered intravenously 5 h after LPS treatment did not reduce histological sign of inflammation, BAL cell recruitment, and protein concentration at 9 h of LPS treatment. We conclude that angiostatin administered subcutaneously after LPS challenge inhibits acute lung inflammation up to 24 h after LPS treatment.


Assuntos
Lesão Pulmonar Aguda/imunologia , Angiostatinas/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Angiostatinas/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Apoptose , Líquido da Lavagem Broncoalveolar , Modelos Animais de Doenças , Injeções Subcutâneas , Lipopolissacarídeos/farmacologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Neutrófilos/imunologia , Peroxidase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Vascul Pharmacol ; 58(4): 280-91, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23220260

RESUMO

Angiostatin is an angiogenesis inhibitor in part generated by and released from platelets. Since platelets upon thrombus formation can give rise to areas of hypoxia, we investigated the effects of angiostatin on endothelial cell migration and apoptosis during hypoxia. Human microvascular endothelial cells (HMVEC-L) were exposed to angiostatin under normoxic or hypoxic conditions. Apoptosis was measured by flow-cytometry. HMVEC-L migration was studied using a modified Boyden Chamber assay, in which migration is MMP-dependent. MMP-2, MMP-14, and VEGF levels were measured using immunoblot, Q-PCR and ELISA. During hypoxia HMVEC-L were protected from angiostatin-induced apoptosis due to increased hypoxia-induced VEGF expression. However, MMP-dependent migration of HMVEC-L was inhibited by angiostatin under hypoxic but not normoxic conditions. Angiostatin decreased MMP-2 at the gene and protein levels only in HMVEC-L exposed to hypoxia. A similar result was obtained for MMP-14. Higher angiostatin concentrations, as would be seen during thrombosis, induced HMVEC-L apoptosis, which was not rescued by VEGF. Under hypoxic conditions angiostatin's primary anti-angiogenic mechanism is likely inhibition of endothelial cell MMP-dependent endothelial cell migration. Only at higher concentrations does angiostatin induce endothelial cell death. This study identifies a novel angiostatin anti-angiogenesis mechanism that is only triggered under pathological-like conditions.


Assuntos
Inibidores da Angiogênese/metabolismo , Angiostatinas/administração & dosagem , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Inibidores da Angiogênese/administração & dosagem , Angiostatinas/metabolismo , Apoptose/fisiologia , Plaquetas/metabolismo , Hipóxia Celular/fisiologia , Linhagem Celular , Movimento Celular/fisiologia , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Immunoblotting , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trombose/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Vestn Oftalmol ; 128(4): 62-5, 2012.
Artigo em Russo | MEDLINE | ID: mdl-22994111

RESUMO

Increased plasminogen level in tear fluid was found within 28 days and increased plasmin activity in 1-3 and 21 days after alkali burn of cornea, this is the time of cornel ulcers development. Increased plasminogen level and plasmin activity in cornea, conjunctiva and intraocular fluid was found in three days after trauma. Subconjunctival injections of angiostatin K1-4,5 (a product of plasminogen metabolism) during 3 weeks resulted in significant suppression of corneal neovascularization within 14 days and of active branching of the vessels in the following. The use of angiostatin reduced depth and area of corneal ulcers. Obtained data shows the promising potential of development of medications based on angiostatin K1-4,5 for suppression of corneal neovascularization and for treatment of diseases associated with corneal ulceration.


Assuntos
Angiostatinas , Córnea/metabolismo , Neovascularização da Córnea/metabolismo , Úlcera da Córnea , Queimaduras Oculares , Neovascularização Patológica/metabolismo , Fluxo Sanguíneo Regional/efeitos dos fármacos , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/metabolismo , Inibidores da Angiogênese/farmacologia , Angiostatinas/administração & dosagem , Angiostatinas/metabolismo , Angiostatinas/farmacologia , Animais , Humor Aquoso/metabolismo , Túnica Conjuntiva/metabolismo , Córnea/irrigação sanguínea , Córnea/fisiopatologia , Neovascularização da Córnea/etiologia , Neovascularização da Córnea/fisiopatologia , Úlcera da Córnea/tratamento farmacológico , Úlcera da Córnea/etiologia , Úlcera da Córnea/metabolismo , Úlcera da Córnea/fisiopatologia , Descoberta de Drogas , Queimaduras Oculares/induzido quimicamente , Queimaduras Oculares/complicações , Queimaduras Oculares/fisiopatologia , Modelos Animais , Neovascularização Patológica/etiologia , Neovascularização Patológica/fisiopatologia , Plasminogênio/metabolismo , Coelhos
4.
Hum Gene Ther ; 22(5): 523-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21443427

RESUMO

Age-related macular degeneration (AMD) is a complex disease that has two phases: a degenerative phase often referred to as nonneovascular AMD (non-NVAMD) or dry AMD and a phase dominated by growth of new blood vessels in the subretinal space, referred to as NVAMD or wet AMD. Advances in the understanding of the molecular pathogenesis of NVAMD have led to new drug therapies that have provided major benefits to patients. However, those treatments require frequent intraocular injections that in many patients must be continued indefinitely to maintain visual benefits. Gene transfer to augment expression of endogenous antiangiogenic proteins is an alternative approach that has the potential to provide long-term stability in patients with NVAMD. Studies in animal models that mimic aspects of NVAMD have identified several possible transgenes, and a clinical trial in patients with advanced NVAMD has suggested that the approach may be feasible. Many important questions remain, but the rationale and preliminary data are compelling. The results of two ongoing clinical trials may answer several of the questions and help direct future research.


Assuntos
Inibidores da Angiogênese/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Terapia Genética/métodos , Degeneração Macular Exsudativa/fisiopatologia , Degeneração Macular Exsudativa/terapia , Angiostatinas/administração & dosagem , Angiostatinas/farmacologia , Dependovirus , Endostatinas/administração & dosagem , Endostatinas/farmacologia , Proteínas do Olho/administração & dosagem , Proteínas do Olho/farmacologia , Vetores Genéticos , Humanos , Fatores de Crescimento Neural/administração & dosagem , Fatores de Crescimento Neural/farmacologia , Serpinas/administração & dosagem , Serpinas/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/administração & dosagem , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/farmacologia
5.
Brain Res ; 1390: 59-69, 2011 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-21443868

RESUMO

The development of the cancer stem cell (CSCs) niche theory has provided a new target for the treatment of gliomas. Gene therapy using oncolytic viral vectors has shown great potential for the therapeutic targeting of CSCs. To explore whether a viral vector carrying an exogenous Endo-Angio fusion gene (VAE) can infect and kill glioma stem cells (GSCs), as well as inhibit their vascular niche in vitro, we have collected surgical specimens of human high-grade glioma (world health organization, WHO Classes III-VI) from which we isolated and cultured GSCs under conditions originally designed for the selective expansion of neural stem cells. Our results demonstrate the following: (1) Four lines of GSCs (isolated from 20 surgical specimens) could grow in suspension, were multipotent, had the ability to self-renew and expressed the neural stem cell markers, CD133 and nestin. (2) VAE could infect GSCs and significantly inhibit their viability. (3) The Endo-Angio fusion gene was expressed in GSCs 48 h after VAE infection and could inhibit the proliferation of human brain microvascular endothelial cells (HBMEC). (4) Residual viable cells lose the ability of self-renewal and adherent differentiation. In conclusion, VAE can significantly inhibit the activity of GSCs in vitro and the expression of exogenous Endo-Angio fusion gene can inhibit HBMEC proliferation. VAE can be used as a novel virus-gene therapy strategy for glioma.


Assuntos
Angiostatinas/genética , Endostatinas/genética , Glioma/genética , Células-Tronco Neoplásicas , Vírus Oncolíticos/genética , Angiostatinas/administração & dosagem , Animais , Sobrevivência Celular/genética , Células Cultivadas , Endostatinas/administração & dosagem , Terapia Genética/métodos , Terapia Genética/tendências , Glioma/patologia , Glioma/terapia , Humanos , Células-Tronco Neoplásicas/patologia , Proteínas de Fusão Oncogênica/administração & dosagem , Proteínas de Fusão Oncogênica/genética , Ratos , Células Tumorais Cultivadas
6.
Biol Reprod ; 84(3): 505-13, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20980690

RESUMO

During placentation, the concentration of fibrinous deposits on the surfaces of maternal vasculature plays a role in villous development and has been strongly implicated in the pathophysiology of human fetal growth restriction (FGR). Fibrinous deposits are conspicuous sites of platelet aggregation where there is local activation of the hemostatic cascade. During activation of the hemostatic cascade, a number of pro- and antiangiogenic agents may be generated at the cell surface, and an imbalance in these factors may contribute to the placental pathology characteristic of FGR. We tested the hypothesis that angiostatin(4.5) (AS(4.5)), a cleavage fragment of plasminogen liberated at the cell surface, is capable of causing FGR in mice. Increased maternal levels of AS(4.5) in vivo result in reproducible placental pathology, including an altered vascular compartment (both in decidual and labyrinthine layers) and increased apoptosis throughout the placenta. In addition, there is significant skeletal growth delay and conspicuous edema in fetuses from mothers that received AS(4.5). Maternally generated AS(4.5), therefore, can access maternal placental vasculature and have a severe effect on placental architecture and inhibit fetal development in vivo. These findings strongly support the hypothesis that maternal AS(4.5) levels can influence placental development, possibly by directly influencing trophoblast turnover in the placenta, and contribute to fetal growth delay in mice.


Assuntos
Angiostatinas/administração & dosagem , Angiostatinas/efeitos adversos , Doenças do Desenvolvimento Ósseo/induzido quimicamente , Doenças Fetais/induzido quimicamente , Doenças Placentárias/induzido quimicamente , Trombofilia/induzido quimicamente , Animais , Doenças do Desenvolvimento Ósseo/patologia , Feminino , Doenças Fetais/patologia , Retardo do Crescimento Fetal/induzido quimicamente , Retardo do Crescimento Fetal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mães , Doenças Placentárias/patologia , Placentação/efeitos dos fármacos , Gravidez , Complicações Hematológicas na Gravidez/induzido quimicamente , Complicações Hematológicas na Gravidez/patologia , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/patologia , Trombofilia/patologia
7.
Cancer Res ; 67(12): 5789-97, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17575146

RESUMO

Antiangiogenic therapy is a promising alternative for prostate cancer growth and metastasis and holds great promise as an adjuvant therapy. The present study evaluated the potential of stable expression of angiostatin and endostatin before the onset of neoplasia and during the early and late stages of prostate cancer progression in transgenic adenocarcinoma of mouse prostate (TRAMP) mice. Groups of 5-, 10-, and 18-week-old male TRAMP mice received recombinant adeno-associated virus-6 encoding mouse endostatin plus angiostatin (E+A) by i.m. injection. The effects of therapy were determined by sacrificing groups of treated mice at defined stages of tumor progression and following cohorts of similarly treated mice for long-term survival. Results indicated remarkable survival after recombinant adeno-associated virus-(E+A) therapy only when the treatment was given at an earlier time, before the onset of high-grade neoplasia, compared with treatment given for invasive cancer. Interestingly, early-stage antiangiogenic therapy arrested the progression of moderately differentiated carcinoma to poorly differentiated state and distant metastasis. Immunohistochemical analysis of the prostate from treated mice indicated significantly lower endothelial cell proliferation and increased tumor cell apoptosis. Vascular endothelial growth factor receptor (VEGFR)-2 expression was significantly down-regulated in tumor endothelium after treatment but not VEGFR-1. Analysis of the neuroendocrine marker synaptophysin expression indicated that antiangiogenic therapy given at an early-stage disease reduced neuroendocrine transition of the epithelial tumors. These studies indicate that stable endostatin and angiostatin gene therapy may be more effective for minimally invasive tumors rather than advanced-stage disease.


Assuntos
Adenocarcinoma/terapia , Inibidores da Angiogênese/administração & dosagem , Angiostatinas/administração & dosagem , Endostatinas/administração & dosagem , Neoplasias da Próstata/terapia , Adenoviridae/genética , Angiostatinas/genética , Animais , Modelos Animais de Doenças , Endostatinas/genética , Ensaio de Imunoadsorção Enzimática , Terapia Genética/métodos , Vetores Genéticos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinaptofisina/efeitos dos fármacos , Sinaptofisina/metabolismo
8.
Ann Oncol ; 17(1): 97-103, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16282244

RESUMO

BACKGROUND: Recombinant human angiostatin (rhAngiostatin) functions as a potent inhibitor of angiogenesis. This study combined rhAngiostatin with a standard chemotherapy regimen in patients with advanced non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS: Eligible patients had chemotherapy-naïve stage IIIB (with pleural effusion) or IV NSCLC, performance status (PS) 0 or 1, no history of bleeding, brain metastasis or requirements for anti-coagulation. Patients received carboplatin (AUC 5) intravenously and paclitaxel (175 mg/m2) intravenously day 1 + subcutaneous rhAngiostatin at either 15 mg or 60 mg twice daily. Cycles were repeated every 3 weeks, for up to six cycles. Patients without progression after completing at least four cycles were continued on maintenance rhAngiostatin until disease progression. RESULTS: Patient characteristics (n = 24) were: 16 males, median age 66 years (range 45-78), 54% PS 1, 83.3% stage IV and 62.5% adenocarcinoma. Grade 3/4 toxicities included: fatigue 47.8%, neutropenia 39.1%, dyspnea 39.1%, vascular 26.1% and infection 17.4%. The overall response rate was 39.1%, 39.1% stable disease and 21.7% progressive disease. Median time to progression was 144 days, and 1-year survival was 45.8%. CONCLUSIONS: rhAngiostatin in combination with paclitaxel and carboplatin is feasible and results in a high disease control rate in patients with advanced NSCLC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Idoso , Angiostatinas/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Carboplatina/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Progressão da Doença , Intervalo Livre de Doença , Estudos de Viabilidade , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Paclitaxel/administração & dosagem , Proteômica , Taxa de Sobrevida , Fatores de Tempo
9.
Lymphology ; 38(3): 146-55, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16353492

RESUMO

Malignant cancers commonly invade locally followed by spread through venous or lymphatic channels or both to distant sites. Hemangiogenesis and its relation to tumor growth and metastasis have been extensively studied. However, the role played by lymphangiogenesis in growth and metastasis of cancer has been largely neglected until just recently. Inhibition of lymphangiogenesis, as compared to inhibition of hemangiogenesis, may provide new insights into the mechanisms of cancer metastasis. The current study was designed to examine the in vitro effect of two commonly used inhibitors of hemangiogenesis, angiostatin and thalidomide, on the growth and proliferation of lymphatic endothelial cells isolated from pig thoracic ducts. We first isolated and characterized the lymphatic endothelial (LE) cells using specific markers for VEGFR3 and LYVE-1. The experimental results showed that treatment of the LE cells with these two drugs resulted in a decrease in the rate of cell proliferation in a dose-dependent manner as assessed by MTT assays. Cell migration rate was assessed by the speed of cell migration from the scrape-wound margin, and the results showed that migration of LE cells was also significantly inhibited in a dose-dependent fashion compared to controls. Treatment with angiostatin and thalidomide both resulted in an increase in apoptosis of LE cells as assessed by Hoechst staining and flow cytometry. We conclude that both angiostatin and thalidomide are able to inhibit LE cell growth in a dose-dependent manner and that the inhibition may be through induction of apoptosis.


Assuntos
Inibidores da Angiogênese/farmacologia , Angiostatinas/farmacologia , Linfangiogênese/efeitos dos fármacos , Talidomida/farmacologia , Inibidores da Angiogênese/administração & dosagem , Angiostatinas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endotélio Linfático/citologia , Endotélio Linfático/efeitos dos fármacos , Citometria de Fluxo , Técnicas In Vitro , Coloração e Rotulagem , Suínos , Talidomida/administração & dosagem
10.
Gynecol Oncol ; 98(2): 203-10, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15975645

RESUMO

OBJECTIVE: To determine the impact of thalidomide and angiostatin on tumor growth, angiogenesis, and apoptosis in a xenograft model of cervical cancer. METHODS: Human umbilical endothelial cells were treated with angiostatin or thalidomide and bFGF-induced proliferation was assessed with the MTT assay. Human cervical cancer cells (CaSki and SiHa) were injected into the flanks of nude mice. After tumors developed, mice were treated with angiostatin 20 mg/kg/day or thalidomide 200 mg/kg/day for 30 days. Fractional tumor growth was determined and immunohistochemical analysis of tumors was used to determine degree of angiogenesis. TUNEL assay was used to assess apoptosis. RESULTS: Angiostatin inhibited endothelial cell proliferation by 50-60%. Thalidomide had no direct effect on endothelial cells. Angiostatin and thalidomide both inhibited tumor growth by about 55%. We found no additive or synergistic effect when the two agents were combined. Both agents inhibited angiogenesis and induced apoptosis when compared to tumors from control animals. CONCLUSIONS: Angiostatin and thalidomide inhibit tumor growth, angiogenesis, and induce apoptosis in this xenograft model of cervical cancer.


Assuntos
Inibidores da Angiogênese/farmacologia , Angiostatinas/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Talidomida/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Inibidores da Angiogênese/administração & dosagem , Angiostatinas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Distribuição Aleatória , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Talidomida/administração & dosagem , Neoplasias do Colo do Útero/irrigação sanguínea , Neoplasias do Colo do Útero/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Int J Colorectal Dis ; 20(6): 494-501, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15864607

RESUMO

METHODS: We applied an experimental approach employing two recombinant adenoviral vectors (Ad) that express interleukin-12 (IL-12) and angiostatin-like molecule (K1-3) respectively to a subcutaneous hepatoma model in mice. RESULTS: Injection of AdK1-3 into tumour nodules established by subcutaneous (s.c.) implantation of Hepa129 hepatoma cells in C3H mice resulted in a significant dose-dependent reduction in tumour growth by 57% in the high dosage group (5x10(9) plaque-forming units [pfu], n=8) 10 days after treatment initiation. Similar antitumoural effects were found for the intratumoural mono-therapy with IL-12 (2.5x10(9) pfu, n=8) resulting in 60% tumour inhibition at the same time point. The survival rate was significantly (p=0.009) improved in the IL-12 but not in the K1-3 treatment group. A combination therapy of AdK1-3 and AdIL-12 was also effective, but did not further improve antitumour efficacy compared with the monotherapy. CONCLUSION: In conclusion, both mono- and combination therapy of K1-3 and IL-12 significantly inhibited tumour progression in this experimental tumour model. The co-administration of both compounds did not result in additive antitumour effects. We hypothesise that the lack of additive antitumour effects of the combination treatment might be attributed to partially counteracting antitumour effects and further studies are needed to illustrate the interference of tumour angiogenesis and tumour inflammation in this tumour model.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Angiostatinas/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/terapia , Terapia Genética/métodos , Interleucina-12/uso terapêutico , Neoplasias Hepáticas Experimentais/terapia , Adenoviridae/genética , Adjuvantes Imunológicos/administração & dosagem , Angiostatinas/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Feminino , Vetores Genéticos , Injeções Intralesionais , Interleucina-12/administração & dosagem , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Resultado do Tratamento
12.
Mol Vis ; 10: 987-95, 2004 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-15623988

RESUMO

PURPOSE: To investigate the effect of different doses of adjuvant angiostatin affecting hepatic micrometastasis in a murine model of metastatic ocular melanoma. METHODS: Angiostatin and plasminogen expression was detected in three murine melanoma cell lines (Queens, B16F10, and B16LS9). The three cell lines were heterotopically inoculated into the posterior compartment (PC) of the right eyes of C57BL/6 mice. After enucleation, the mice were given injections of 100 microl PBS and low dose (0.1 microg/microl) or high dose (0.3 microg/microl) murine recombinant angiostatin every day for 14 days after enucleation. The mice were sacrificed at 21 days post-enucleation and hepatic micrometastases were counted. In vitro migration/invasion assays were performed with low (0.1 microg/microl) and high (50 microg/microl) concentration angiostatin supplementation. Quantitative RT-PCR detected mRNA and Western analysis determined protein expression of VEGF for all cell lines. Evaluation of TdT mediated dUTP nick end labeling (TUNEL) and MIB1 immunostaining of the micrometastases determined apoptosis and proliferation ratios. RESULTS: There was a decrease in micrometastasis in the low dose group for Queens (p<0.05), B16F10 (p<0.05), and B16LS9 melanoma (p<0.01) cell lines. Two of the cell lines (B16F10 and B16LS9) elaborated plasminogen and were able to cleave plasminogen into K1-K4 (angiostatin). There was a decrease in the in vitro migration and invasion after supplementation with low concentration compared to high concentration angiostatin (p<0.01). VEGF mRNA and protein expression decreased in all cells lines in low concentration angiostatin, with the greatest decrease in B16LS9 cells (p<0.05). Apoptosis ratios were increased (p<0.01) and proliferation ratios were decreased (p<0.01) in hepatic micrometastases after treatment with low dose angiostatin. CONCLUSIONS: There were significantly fewer micrometastases in treated compared to controls with low dose compared to high dose angiostatin. This treatment results in apoptosis in the micrometastases. The mechanism appears to be related an anti-migratory effect and altered VEGF expression by melanoma cells.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Angiostatinas/administração & dosagem , Antineoplásicos/administração & dosagem , Neoplasias Oculares/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Animais , Apoptose , Western Blotting , Proliferação de Células , Modelos Animais de Doenças , Enucleação Ocular , Neoplasias Oculares/genética , Neoplasias Oculares/patologia , Feminino , Marcação In Situ das Extremidades Cortadas , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundário , Melanoma Experimental/genética , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , Plasminogênio/genética , Plasminogênio/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
J Vasc Interv Radiol ; 15(6): 601-8, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15178721

RESUMO

PURPOSE: To evaluate the importance of angiogenesis in plaque progression after stent placement, this study examines stent-based controlled delivery of the antiangiogenic agent, angiostatin, in a rabbit model. MATERIALS AND METHODS: Controlled release biodegradable microspheres delivering angiostatin or polymer-only microspheres (polylactic-co-glycolic-acid-polyethylene glycol; PLGA/PEG) were loaded in channeled stents, anchored, and deployed in the aorta of adult New Zealand white rabbits (n = 6 animals per group, three each per time point). The stented aortas were harvested at 7 days and 28 days and evaluated for neovascularization, local inflammation, vascular smooth muscle cell proliferation, and in-stent plaque progression. RESULTS: At 7 days, neovascularization was significantly decreased in the angiostatin groups (1.6 +/- 1.6 neovessels per mm(2) plaque) versus the control group (15.4 +/- 2.6 neovessels per mm(2) plaque; P =.00081), as were local inflammation where angiostatin-treated groups demonstrated significantly lower macrophage recruitment per cross section (34.9 +/- 4.9 cells per cross section) relative to the control group (55.2 +/- 3.84 cells per cross section; P =.0037). And a significant decrease in the overall vascular smooth muscle cell proliferation (143.8 +/- 26.3 Ki-67 positive cells per mm(2)) relative to the control group (263.2 +/- 16.6 Ki-67 positive cells per mm(2); P =.00074). At both 7 and 28 days, in-stent plaque progression in the angiostatin groups was successfully limited relative to the control group by 54% (0.255 +/- 0.019% of cross section; P =.00016) and 19% (1.981 +/- 0.080; P =.0033) respectively and resulted in reduction of in-stent restenosis relative to the control group. CONCLUSION: Angiostatin-eluting stents may limit neovascularity after arterial implantation, offer insight into in-stent restenosis, and allow future refinement of bioactive stent designs and clinical strategies, particularly in light of evidence that intimal smooth muscle cells may in part be marrow-derived.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Angiostatinas/administração & dosagem , Aorta Abdominal , Arteriopatias Oclusivas/patologia , Materiais Revestidos Biocompatíveis , Oclusão de Enxerto Vascular/prevenção & controle , Stents , Análise de Variância , Animais , Aorta Abdominal/patologia , Géis , Masculino , Microesferas , Coelhos
14.
Microvasc Res ; 67(2): 105-16, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15020201

RESUMO

We present a method whereby en face estimation of the chorionic capillary plexus can be generated in the living chick chorioallantoic membrane (CAM) and confirmed by post-fixation cross section analysis. This value does not alter significantly with age and provides a reliable and simple method to evaluate anti-angiogenesis. Anti-angiogenesis may be induced by an intervention, such as a pharmacological agent, applied to the surface of the CAM. We describe the use of silastic rings that are associated with minimal inflammatory reaction, in this process. By estimating changes in the chorionic capillary plexus to quantify anti-angiogenesis, together with silastic rings, we examined the anti-angiogenic effect of human angiostatin and demonstrated that although there is a significant loss of capillaries en face after exposure from days 7 to 9 of incubation, in contrast there is no significant inhibition after exposure to a similar dose of angiostatin from days 11 to 13 of incubation. This not only demonstrates the important effects on neo-angiogenesis compared to mature vessels, but also illustrates the potential of the CAM to readily provide a means for such a comparison.


Assuntos
Alantoide/efeitos dos fármacos , Angiostatinas/farmacologia , Córion/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Fatores Etários , Alantoide/irrigação sanguínea , Angiostatinas/administração & dosagem , Animais , Capilares/efeitos dos fármacos , Embrião de Galinha , Córion/irrigação sanguínea , Dimetilpolisiloxanos , Portadores de Fármacos , Implantes de Medicamento , Glucose/farmacologia , Humanos , Microscopia de Vídeo , Silicones , Soluções/farmacologia
15.
J Cancer Res Clin Oncol ; 130(1): 15-24, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14586644

RESUMO

Tumor cells secrete diffusible substances collectively called tumor angiogenic factors (TAFs), most notably vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), which in turn stimulate endothelial cell migration and thus angiogenesis, or new blood vessel formation. Anti-angiogenic drugs for cancer treatment are receiving much attention, with endostatin identified as one of the potent inhibitors. Although the mechanisms of action of endostatin are yet to be fully elucidated, there is evidence that bFGF and endostatin may bind competitively to heparan sulfate proteoglycan receptors on endothelial cells, or endostatin may otherwise downregulate bFGF or VEGF and its receptors, putatively inhibiting cell proliferation. To test these and other hypotheses of inhibitory action that can be similarly formulated, for other TAF inhibitors as well as endostatin, we have developed a mathematical model of extratumoral angiogenesis in cancer in response to specific anti-angiogenic drug treatment. It is built on previous work, a modification and augmentation of published models, and is expressed as four nonlinear partial differential equations, with specific terms for endothelial cell proliferation, degradation, and endostatin-TAF inhibition, and a stochastic, discretized version of this model to represent vessel growth. Our extended model reproduces the simulated kinetics of angiogenesis in a mouse tumor model reported earlier. We assessed the anti-angiogenic kinetic behavior of our extended model by simulating dynamic responses to exogenous endostatin treatment in the same mouse model, using four dosage regimens, two of these reported for in vivo pre-clinical or clinical studies, and two 10 times greater: daily single bolus injections of 20 mg/kg per day and 200 mg/kg per day, and constant infusions of 20 mg/kg per day and 200 mg/kg per day, each for 20 simulated days. We also explored the effects of drug clearance, over an eightfold range of clearance rates that include scaled clearances for endostatin, a sister-drug angiostatin, or similar drugs with clearances in this range. Predictively, our simulation results suggest ineffectiveness of the bolus injection protocols, consistent with in vivo data with angiostatin treatment, whereas simulated constant infusion of endostatin in the mouse model effectively suppresses angiogenesis after only 3 days of treatment, at the lowest dose, over a wide range of drug clearance rates.


Assuntos
Inibidores da Angiogênese/farmacologia , Angiostatinas/farmacologia , Endostatinas/farmacologia , Neovascularização Patológica , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacocinética , Angiostatinas/administração & dosagem , Angiostatinas/farmacocinética , Animais , Vias de Administração de Medicamentos , Esquema de Medicação , Endostatinas/administração & dosagem , Endostatinas/farmacocinética , Humanos , Computação Matemática , Modelos Teóricos , Neovascularização Patológica/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...