Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clin Immunol ; 163: 84-90, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26769276

RESUMO

Complement activation contributes to inflammation in many diseases, yet it also supports physiologic apoptotic cells (AC) clearance and its downstream immunosuppressive effects. The roles of individual complement components in AC phagocytosis have been difficult to dissect with artificially depleted sera. Using human in vitro systems and the novel antibody complement C1s inhibitor TNT003, we uncoupled the role of the enzymatic activation of the classical pathway from the opsonizing role of C1q in mediating a) the phagocytosis of early and late AC, and b) the immunosuppressive capacity of early AC. We found that C1s inhibition had a small impact on the physiologic clearance of early AC, leaving their immunosuppressive properties entirely unaffected, while mainly inhibiting the phagocytosis of late apoptotic/secondary necrotic cells. Our data suggest that C1s inhibition may represent a valuable therapeutic strategy to control classical pathway activation without causing significant AC accumulation in diseases without defects in AC phagocytosis.


Assuntos
Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Complemento C1q/efeitos dos fármacos , Complemento C1s/antagonistas & inibidores , Citocinas/efeitos dos fármacos , Citofagocitose/efeitos dos fármacos , Tolerância Imunológica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Apoptose/imunologia , Ativação do Complemento/imunologia , Complemento C1q/imunologia , Complemento C1q/metabolismo , Complemento C1s/metabolismo , Complemento C3b/efeitos dos fármacos , Complemento C3b/imunologia , Complemento C3b/metabolismo , Citocinas/imunologia , Citofagocitose/imunologia , Humanos , Técnicas In Vitro , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Células Jurkat , Macrófagos/imunologia , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/imunologia
2.
Blood ; 123(26): 4015-22, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24695853

RESUMO

Activation of the classical pathway (CP) of complement is often associated with autoimmune disorders in which disease pathology is linked to the presence of an autoantibody. One such disorder is cold agglutinin disease (CAD), an autoimmune hemolytic anemia in which autoantibodies (cold agglutinins) bind to red blood cells (RBCs) at low temperatures. Anemia occurs as a result of autoantibody-mediated CP activation on the surface of the erythrocyte, leading to the deposition of complement opsonins that drive extravascular hemolysis in the liver. Here we test the effects of TNT003, a mouse monoclonal antibody targeting the CP-specific serine protease C1s, on CP activity induced by cold agglutinins on human RBCs. We collected 40 individual CAD patient samples and showed that TNT003 prevented cold agglutinin-mediated deposition of complement opsonins that promote phagocytosis of RBCs. Furthermore, we show that by preventing CP activation, TNT003 also prevents cold agglutinin-driven generation of anaphylatoxins. Finally, we provide evidence that CP activity in CAD patients terminates prior to activation of the terminal cascade, supporting the hypothesis that the primary route of RBC destruction in these patients occurs via extravascular hemolysis. Our results support the development of a CP inhibitor for the treatment of CAD.


Assuntos
Anemia Hemolítica Autoimune/tratamento farmacológico , Anticorpos Monoclonais Murinos/farmacologia , Ativação do Complemento/efeitos dos fármacos , Complemento C1s/antagonistas & inibidores , Serina Proteases/sangue , Inibidores de Serina Proteinase/farmacologia , Idoso , Anafilatoxinas/metabolismo , Anemia Hemolítica Autoimune/enzimologia , Anemia Hemolítica Autoimune/patologia , Animais , Linhagem Celular Tumoral , Complemento C1s/metabolismo , Feminino , Hemólise/efeitos dos fármacos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade
3.
Cancer Res ; 73(11): 3316-25, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23667176

RESUMO

LD22-4, an 86-amino acid fragment of the basic fibroblast growth factor, is an inhibitor of cell migration. LD22-4 inhibits the migration of various tumor cells, endothelial cells, and fibroblasts in vitro and suppresses tumor growth and angiogenesis in vivo. LD22-4 is effective in the presence of multiple growth factors, either alone or in combination, as well as haptotactic factors. LD22-4 inhibits the rate of malignant gliomas prepared from U87MG cells in an orthotopic mouse model by 90% compared with untreated mice. Using U87MG cells, we identified the LD22-4 membrane receptor as neuropilin 1 (NRP1). The identification of NRP1 as the LD22-4 receptor was based upon mass spectrometric analysis of proteins that bind to LD22-4, immunoprecipitation of an NRP1-LD22-4 complex formed during incubation of LD22-4 with U87MG cells, LD22-4-NRP1 coimmunoprecipitation studies, and binding of LD22-4 to HEK293 cells expressing NRP1. In contrast, NRP1 binding of an inactive mutant of LD22-4 was substantially reduced. As is typical of NRP1-binding proteins, LD22-4 itself binds to heparin and requires heparan sulfate for binding to cells. The addition of heparin to migration assays increased the inhibitory activity of LD22-4. In addition to a heparin-binding region, LD22-4 contains a 5-amino acid C-terminus that matches an NRP1 consensus binding sequence. Thus, direct binding experiments, dependence on heparan sulfate, and the presence of a NRP1 consensus binding sequence indicate that NRP1 is the binding site of LD22-4 and mediates inhibition of cell migration.


Assuntos
Movimento Celular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Glioma/tratamento farmacológico , Neuropilina-1/metabolismo , Fragmentos de Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Processos de Crescimento Celular/efeitos dos fármacos , Feminino , Fator 2 de Crescimento de Fibroblastos/química , Glioma/metabolismo , Glioma/patologia , Células HEK293 , Heparina/química , Heparina/metabolismo , Humanos , Camundongos , Camundongos Nus , Neuropilina-1/antagonistas & inibidores , Fragmentos de Peptídeos/química , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Clin Cancer Res ; 14(7): 2137-44, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18381955

RESUMO

PURPOSE: ATN-161 (Ac-PHSCN-NH(2)) is an integrin-binding peptide that is currently in phase II trials in cancer patients. This peptide has been shown to have antitumor activity in a number of different preclinical models. EXPERIMENTAL DESIGN: In this study, we examined the binding, biodistribution, and dose and biomarker response of ATN-161 in several animal models. RESULTS: ATN-161 bound to the beta subunit of a number of different integrins implicated in tumor growth and progression, which depended on its cysteine thiol. The peptide had antiangiogenic activity in the Matrigel plug model, and this activity could be reversed by inhibitors of protein kinase A, an effector of alpha(5)beta(1)-dependent angiogenesis. A labeled analogue of ATN-161, ATN-453, localized to neovessels but not to preexisting vasculature in vivo. The half-life of the peptide when localized to a tumor was much longer than in plasma. Dose-response studies in the Matrigel plug model of angiogenesis or a Lewis lung carcinoma model of tumor growth showed a U-shaped dose-response curve with 1 to 10 mg/kg given thrice a week, being the optimal dose range of ATN-161. Two additional pharmacodynamic models of angiogenesis (dynamic contrast-enhanced magnetic resonance imaging and measurement of endothelial cell progenitors) also revealed U-shaped dose-response curves. CONCLUSIONS: The presence of a U-shaped dose-response curve presents a significant challenge to identifying a biologically active dose of ATN-161. However, the identification of biomarkers of angiogenesis that also exhibit this same U-shaped response should allow the translation of those biomarkers to the clinic, allowing them to be used to identify the active dose of ATN-161 in phase II studies.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Oligopeptídeos/farmacologia , Animais , Biomarcadores Tumorais/análise , Relação Dose-Resposta a Droga , Humanos , Camundongos , Neovascularização Patológica/tratamento farmacológico
5.
Mol Cancer Ther ; 5(9): 2271-80, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16985061

RESUMO

PURPOSE: Integrins are expressed by numerous tumor types including breast cancer, in which they play a crucial role in tumor growth and metastasis. In this study, we evaluated the ability of ATN-161 (Ac-PHSCN-NH2), a 5-mer capped peptide derived from the synergy region of fibronectin that binds to alpha5beta1 and alphavbeta3 in vitro, to block breast cancer growth and metastasis. EXPERIMENTAL DESIGN: MDA-MB-231 human breast cancer cells were inoculated s.c. in the right flank, or cells transfected with green fluorescent protein (MDA-MB-231-GFP) were inoculated into the left ventricle of female BALB/c nu/nu mice, resulting in the development of skeletal metastasis. Animals were treated with vehicle alone or by i.v. infusion with ATN-161 (0.05-1 mg/kg thrice a week) for 10 weeks. Tumor volume was determined at weekly intervals and tumor metastasis was evaluated by X-ray, microcomputed tomography, and histology. Tumors were harvested for histologic evaluation. RESULT: Treatment with ATN-161 caused a significant dose-dependent decrease in tumor volume and either completely blocked or caused a marked decrease in the incidence and number of skeletal as well as soft tissue metastases. This was confirmed histologically as well as radiographically using X-ray and microcomputed tomography. Treatment with ATN-161 resulted in a significant decrease in the expression of phosphorylated mitogen-activated protein kinase, microvessel density, and cell proliferation in tumors grown in vivo. CONCLUSION: These studies show that ATN-161 can block breast cancer growth and metastasis, and provides a rationale for the clinical development of ATN-161 for the treatment of breast cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias Ósseas/prevenção & controle , Neoplasias Ósseas/secundário , Neoplasias da Mama/tratamento farmacológico , Oligopeptídeos/farmacologia , Adenocarcinoma/patologia , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Neovascularização Patológica/tratamento farmacológico , Radiografia/instrumentação , Neoplasias de Tecidos Moles/prevenção & controle , Neoplasias de Tecidos Moles/secundário , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Science ; 311(5761): 656-9, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16456079

RESUMO

The urokinase plasminogen activator binds to its cellular receptor with high affinity and initiates signaling cascades that are implicated in pathological processes including tumor growth, metastasis, and inflammation. We report the crystal structure at 1.9 angstroms of the urokinase receptor complexed with the urokinase amino-terminal fragment and an antibody against the receptor. The three domains of urokinase receptor form a concave shape with a central cone-shaped cavity where the urokinase fragment inserts. The structure provides insight into the flexibility of the urokinase receptor that enables its interaction with a wide variety of ligands and a basis for the design of urokinase-urokinase receptor antagonists.


Assuntos
Receptores de Superfície Celular/química , Ativador de Plasminogênio Tipo Uroquinase/química , Anticorpos/química , Anticorpos/metabolismo , Cristalografia por Raios X , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Modelos Moleculares , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
7.
Cancer Res ; 65(17): 7775-81, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16140945

RESUMO

Pancreatic carcinomas express high levels of urokinase-type plasminogen activator (uPA) and its receptor (uPAR), both of which mediate cell migration and invasion. We investigated the hypotheses that (a) insulin-like growth factor-I (IGF-I)- and hepatocyte growth factor (HGF)-mediated migration and invasion of human pancreatic carcinoma cells require uPA and uPAR function and (b) inhibition of uPAR inhibits tumor growth, retroperitoneal invasion, and hepatic metastasis of human pancreatic carcinomas in mice. Using transwell assays, we investigated the effect of IGF-I and HGF on L3.6pl migration and invasion. We measured the induction of uPA and uPAR following treatment of cells with IGF-I and HGF using immunoprecipitation and Western blot analysis. The importance of uPA and uPAR on L3.6pl cell migration and invasion was studied by inhibiting their activities with amiloride and antibodies before cytokine treatment. In an orthotopic mouse model of human pancreatic carcinoma, we evaluated the effect of anti-uPAR monoclonal antibodies with and without gemcitabine on primary tumor growth, retroperitoneal invasion, and hepatic metastasis. IGF-I and HGF mediated cell migration and invasion in L3.6pl cells. In addition, IGF-I and HGF induced uPA and uPAR expression in L3.6pl cells. In vitro, blockade of uPA and uPAR activity inhibited IGF-I- and HGF-mediated cell migration and invasion. Treatment of mice with anti-uPAR monoclonal antibody significantly decreased pancreatic tumor growth and hepatic metastasis and completely inhibited retroperitoneal invasion. Our study shows the importance of the uPA/uPAR system in pancreatic carcinoma cell migration and invasion. These findings suggest that uPAR is a potential target for therapy in patients with pancreatic cancer.


Assuntos
Movimento Celular/fisiologia , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Receptor IGF Tipo 1/antagonistas & inibidores , Receptores de Superfície Celular/antagonistas & inibidores , Amilorida/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Neoplasias Hepáticas Experimentais/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Neovascularização Patológica , Neoplasias Pancreáticas/irrigação sanguínea , Proteínas Proto-Oncogênicas c-met/fisiologia , Receptor IGF Tipo 1/fisiologia , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/imunologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/biossíntese
8.
Ann Surg ; 241(5): 748-56; discussion 756-8, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15849510

RESUMO

OBJECTIVE: To determine whether insulinlike growth factor-I (IGF-I) and hepatocyte growth factor (HGF) cooperate to induce migration and invasion of human colorectal carcinoma (CRC) cells and whether the effects of IGF-I and/or HGF are mediated through activation of the urokinase plasminogen activator (uPA)/uPA receptor (uPAR) system, a central mediator of tumor-cell migration and invasion. SUMMARY BACKGROUND DATA: CRC cells must invade through the basement membrane of the colon and migrate to form metastases. CRC cells are known to overexpress IGF-I receptor (IGF-IR), c-Met, and uPAR, 3 cell-surface receptors known to mediate cell migration and invasion. We hypothesized that IGF-IR and c-Met cooperate to induce migration and invasion in CRC cells and that this signaling is dependent on uPAR. METHODS: KM12L4 human CRC cells were treated with IGF-I, HGF, or IGF-I + HGF in transwell migration and invasion chambers; cells that had migrated or invaded were counted. To determine the role of c-Met in IGF-I-induced migration and invasion, c-Met was inhibited by infection of cells with an adenovirus containing a c-Met ribozyme; transwell assays were then repeated. To determine the role of the uPA/uPAR system in IGF-I-induced CRC cell migration and invasion, transwell assays were repeated after pretreating cells with the uPA inhibitor amiloride or with neutralizing antibodies to uPA and uPAR. RESULTS: IGF-I and HGF, alone or in combination, increased cell migration and invasion. The c-Met ribozyme inhibited IGF-I- and HGF-mediated migration and invasion, indicating that c-Met is essential for these processes. uPA and uPAR inhibition blocked IGF-I- and HGF-mediated migration and invasion, suggesting that uPAR is downstream of IGF/IGF-IR and HGF/c-Met in the signaling pathways that mediate cell migration and invasion. CONCLUSIONS: IGF-I and HGF cooperate to induce migration and invasion of CRC cells, and c-Met and uPA/uPAR are required for IGF-I-mediated migration and invasion. In our in vitro model of CRC migration and invasion, uPA and uPAR appear to be downstream of IGF-IR and c-Met and are required for migration and invasion. Elucidation of the pathways that contribute to tumor progression and metastasis should provide a foundation for the rational development and use of targeted therapies for CRC.


Assuntos
Movimento Celular/fisiologia , Neoplasias Colorretais/patologia , Fator de Crescimento Insulin-Like I/fisiologia , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-met/fisiologia , Receptores de Superfície Celular/fisiologia , Linhagem Celular Tumoral , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transdução de Sinais/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia
9.
Int J Cancer ; 104(4): 496-503, 2003 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-12584749

RESUMO

Integrin alpha(5)beta(1) is expressed on activated endothelial cells and plays a critical role in tumor angiogenesis. We hypothesized that a novel integrin alpha(5)beta(1) antagonist, ATN-161, would inhibit angiogenesis and growth of liver metastases in a murine model. We further hypothesized that combining ATN-161 with 5-fluorouracil (5-FU) chemotherapy would enhance the antineoplastic effect. Murine colon cancer cells (CT26) were injected into spleens of BALB/c mice to produce liver metastases. Four days thereafter, mice were given either ATN-161 (100 mg/kg, every 3rd day) or saline by intraperitoneal injection, with or without combination of continuous-infusion 5-FU (100 mg/kg/2 weeks), which was started on day 7. On day 20 after tumor cell inoculation, mice were killed and liver weights and number of liver metastases were determined. A follow-up study on survival was also conducted in which mice were randomized to receive ATN-161, 5-FU or ATN-161+5-FU. Combination therapy with ATN-161+5-FU significantly reduced tumor burden (liver weight) and number of liver metastases (p<0.02). Liver tumors in the ATN-161 and ATN-161+5-FU groups had significantly fewer microvessels (p<0.05) than tumors in the control or 5-FU-treated groups. Unlike treatment with either agent alone, ATN-161+5-FU significantly increased tumor cell apoptosis and decreased tumor cell proliferation (p<0.03) and improved overall survival (p<0.03, log-rank test). Targeting integrin alpha(5)beta(1) in combination with 5-FU infusion reduced liver metastases formation and improved survival in this colon cancer model. The enhancement of antineoplastic activity from the combination of anti-angiogenic therapy and chemotherapy may be a promising approach for treating metastatic colorectal cancer.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Fluoruracila/administração & dosagem , Integrina alfa5beta1/antagonistas & inibidores , Neoplasias Hepáticas/secundário , Oligopeptídeos/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Integrina alfa5beta1/fisiologia , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C
10.
Cancer Res ; 62(18): 5344-50, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12235005

RESUMO

Histidine-proline-rich glycoprotein (HPRG) is an abundant multidomain plasma protein evolutionarily related to high-molecular-weight kininogen. The cleaved form of high-molecular-weight kininogen has recently been demonstrated to exhibit antiangiogenic activities in vitro (J. C. Zhang et al., FASEB J., 14: 2589-2600, 2000), mediated primarily through domain 5. HPRG contains a histidine-proline-rich (H/P) domain with sequence and functional similarities to HKa-D5. We hypothesized that HPRG may also have antiangiogenic properties, localized within its H/P domain. The H/P domain is highly conserved among species, and because rabbit H/P domain is more resistant to internal proteolytic cleavage than the human domain, the rabbit HPRG (rbHPRG) was primarily used to assess the antiangiogenic activity of HPRG. Rabbit HPRG inhibited human umbilical vein endothelial cell (HUVEC) tube formation stimulated by fibroblast growth factor-2 (FGF-2) or vascular endothelial growth factor on a Matrigel surface as well as cell proliferation of FGF-2 stimulated HUVECs. The antiangiogenic activity of rbHPRG was localized to the H/P domain by use of proteolytic fragments of rbHPRG and was further confirmed and characterized in two in vivo models of angiogenesis: the chorioallantoic membrane of the chick assay and the mouse Matrigel plug assay. Caspase-3 activation was observed in HUVECs stimulated with FGF-2 in the presence of rbHPRG, suggesting that apoptosis of activated endothelial cells may be one of the mechanisms underlying its antiangiogenic activity. Finally, the H/P domain of rbHPRG reduced tumor cell number when tumor cells were co-inoculated in the Matrigel plug assay. In conclusion, the H/P domain within HPRG induces the apoptosis of activated endothelial cells leading to potent antiangiogenic effects.


Assuntos
Inibidores da Angiogênese/farmacologia , Proteínas/farmacologia , Inibidores da Angiogênese/fisiologia , Animais , Divisão Celular/efeitos dos fármacos , Embrião de Galinha , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Fisiológica/efeitos dos fármacos , Plasminogênio/farmacologia , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Estrutura Terciária de Proteína , Proteínas/fisiologia , Coelhos , Sequências Repetitivas de Aminoácidos , Homologia de Sequência de Aminoácidos
11.
J Biol Chem ; 277(34): 31056-61, 2002 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-12063256

RESUMO

The 24-kDa form of basic fibroblast growth factor inhibits the migration of endothelial cells and mammary carcinoma cells while continuing to promote cell proliferation. This molecule consists of the 18-kDa fibroblast growth factor sequence plus an additional 55 amino acids at the amino-terminal end. Antibody neutralization studies suggested that the inhibition of migration is associated with these 55 amino acids, whereas the promotion of proliferation localizes to the 18-kDa domain. To determine whether 24kD basic fibroblast growth factor could be modified to eliminate its effect on cell proliferation but retain its inhibition of migration, portions of the carboxyl-terminal end of 24kD fibroblast growth factor were deleted, and the products were tested on MCF-7 and endothelial cells. A protein consisting of the 55 amino acids of the amino-terminal end and the first 31 amino acids of 18kD basic fibroblast growth factor (ATE+31) inhibited migration by 80% but did not promote cell growth. Arginine to alanine substitutions within the first 21 amino acids of the carboxyl-terminal end substantially reduced the efficacy of ATE+31, whereas substitutions in the remaining part of the molecule had no effect. Competition binding experiments showed that ATE+31 does not compete with 24kD basic fibroblast growth factor for binding to fibroblast growth factor receptor 1. In an in vivo matrigel plug assay, 150 nm ATE+31 peptide reduced angiogenesis by 80%. These studies demonstrate that the amino-terminal end of 24kD basic fibroblast growth factor is responsible for an activity that inhibits the migration rates of cultured cells as well as the angiogenic response in vivo.


Assuntos
Inibidores da Angiogênese/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fragmentos de Peptídeos/farmacologia , Movimento Celular/efeitos dos fármacos , Feminino , Humanos , Peso Molecular , Relação Estrutura-Atividade , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA