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.
Sci Signal ; 8(403): ra117, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26577922

RESUMO

In endothelial cells, binding of vascular endothelial growth factor (VEGF) to the receptor VEGFR2 activates multiple signaling pathways that trigger processes such as proliferation, survival, and migration that are necessary for angiogenesis. VEGF-bound VEGFR2 becomes internalized, which is a key step in the proangiogenic signal. We showed that the urokinase plasminogen activator receptor (uPAR) interacted with VEGFR2 and described the mechanism by which this interaction mediated VEGF signaling and promoted angiogenesis. Knockdown of uPAR in human umbilical vein endothelial cells (HUVECs) impaired VEGFR2 signaling, and uPAR deficiency in mice prevented VEGF-induced angiogenesis. Upon exposure of HUVECs to VEGF, uPAR recruited the low-density lipoprotein receptor-related protein 1 (LRP-1) to VEGFR2, which induced VEGFR2 internalization. Thus, the uPAR-VEGFR2 interaction is crucial for VEGF signaling in endothelial cells.


Assuntos
Neovascularização Fisiológica/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Endocitose , Células Endoteliais da Veia Umbilical Humana , Humanos , Integrina beta1/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Ligação Proteica , Transdução de Sinais
3.
Nat Med ; 20(7): 741-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24929950

RESUMO

The N-terminal fragment of prolactin (16K PRL) inhibits tumor growth by impairing angiogenesis, but the underlying mechanisms are unknown. Here, we found that 16K PRL binds the fibrinolytic inhibitor plasminogen activator inhibitor-1 (PAI-1), which is known to contextually promote tumor angiogenesis and growth. Loss of PAI-1 abrogated the antitumoral and antiangiogenic effects of 16K PRL. PAI-1 bound the ternary complex PAI-1-urokinase-type plasminogen activator (uPA)-uPA receptor (uPAR), thereby exerting antiangiogenic effects. By inhibiting the antifibrinolytic activity of PAI-1, 16K PRL also protected mice against thromboembolism and promoted arterial clot lysis. Thus, by signaling through the PAI-1-uPA-uPAR complex, 16K PRL impairs tumor vascularization and growth and, by inhibiting the antifibrinolytic activity of PAI-1, promotes thrombolysis.


Assuntos
Fibrinólise , Neovascularização Patológica , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Prolactina/fisiologia , Animais , Divisão Celular , Células Cultivadas , Humanos , Camundongos , Camundongos Knockout , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/fisiologia , Prolactina/química
4.
J Clin Invest ; 123(5): 2143-54, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23619365

RESUMO

Peripartum cardiomyopathy (PPCM) is a life-threatening pregnancy-associated cardiomyopathy in previously healthy women. Although PPCM is driven in part by the 16-kDa N-terminal prolactin fragment (16K PRL), the underlying molecular mechanisms are poorly understood. We found that 16K PRL induced microRNA-146a (miR-146a) expression in ECs, which attenuated angiogenesis through downregulation of NRAS. 16K PRL stimulated the release of miR-146a-loaded exosomes from ECs. The exosomes were absorbed by cardiomyocytes, increasing miR-146a levels, which resulted in a subsequent decrease in metabolic activity and decreased expression of Erbb4, Notch1, and Irak1. Mice with cardiomyocyte-restricted Stat3 knockout (CKO mice) exhibited a PPCM-like phenotype and displayed increased cardiac miR-146a expression with coincident downregulation of Erbb4, Nras, Notch1, and Irak1. Blocking miR-146a with locked nucleic acids or antago-miRs attenuated PPCM in CKO mice without interrupting full-length prolactin signaling, as indicated by normal nursing activities. Finally, miR-146a was elevated in the plasma and hearts of PPCM patients, but not in patients with dilated cardiomyopathy. These results demonstrate that miR-146a is a downstream-mediator of 16K PRL that could potentially serve as a biomarker and therapeutic target for PPCM.


Assuntos
Cardiomiopatias/sangue , Cardiomiopatias/genética , MicroRNAs/sangue , Complicações Cardiovasculares na Gravidez/sangue , Prolactina/metabolismo , Animais , Biomarcadores/sangue , Células Endoteliais/citologia , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Neovascularização Patológica , Período Periparto , Gravidez , Complicações Cardiovasculares na Gravidez/metabolismo , Ratos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
5.
PLoS One ; 6(11): e27318, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22087289

RESUMO

BACKGROUND: Angiogenesis, the formation of new blood vessels from existing vasculature, plays an essential role in tumor growth, invasion, and metastasis. 16K hPRL, the antiangiogenic 16-kDa N-terminal fragment of human prolactin was shown to prevent tumor growth and metastasis by modifying tumor vessel morphology. METHODOLOGY/PRINCIPAL FINDINGS: Here we investigated the effect of 16K hPRL on tumor vessel maturation and on the related signaling pathways. We show that 16K hPRL treatment leads, in a murine B16-F10 tumor model, to a dysfunctional tumor vasculature with reduced pericyte coverage, and disruption of the PDGF-B/PDGFR-B, Ang/Tie2, and Delta/Notch pathways. In an aortic ring assay, 16K hPRL impairs endothelial cell and pericyte outgrowth from the vascular ring. In addition, 16K hPRL prevents pericyte migration to endothelial cells. This event was independent of a direct inhibitory effect of 16K hPRL on pericyte viability, proliferation, or migration. In endothelial cell-pericyte cocultures, we found 16K hPRL to disturb Notch signaling. CONCLUSIONS/SIGNIFICANCE: Taken together, our data show that 16K hPRL impairs functional tumor neovascularization by inhibiting vessel maturation and for the first time that an endogenous antiangiogenic agent disturbs Notch signaling. These findings provide new insights into the mechanisms of 16K hPRL action and highlight its potential for use in anticancer therapy.


Assuntos
Inibidores da Angiogênese/farmacologia , Vasos Sanguíneos/crescimento & desenvolvimento , Neoplasias/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Prolactina/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Técnicas de Cocultura , Células Endoteliais , Camundongos , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Pericitos , Prolactina/uso terapêutico , Transdução de Sinais/efeitos dos fármacos
6.
Cancer Lett ; 284(2): 222-8, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19473755

RESUMO

Human 16K PRL (16K hPRL) is a potent inhibitor of angiogenesis both in vitro and in vivo. It has been shown to prevent tumor growth in three xenograft mouse models. Here we have used a gene transfer method based on cationic liposomes to produce 16K hPRL and demonstrate that 16K hPRL inhibits tumor growth in a subcutaneous B16F10 mouse melanoma model. Computer-assisted image analysis shows that 16K hPRL treatment results in the reduction of tumor vessel length and width, leading to a 57% reduction in average vessel size. We thus show, for the first time, that administration of the 16K hPRL gene complexed to cationic liposomes is effective to maintain antiangiogenic activities of 16K hPRL level.


Assuntos
Inibidores da Angiogênese/uso terapêutico , DNA Recombinante/uso terapêutico , Terapia Genética , Melanoma Experimental/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Prolactina/uso terapêutico , Inibidores da Angiogênese/administração & dosagem , Animais , Cátions , Linhagem Celular , Colesterol , DNA Recombinante/administração & dosagem , Ensaios de Seleção de Medicamentos Antitumorais , Ácidos Graxos Monoinsaturados , Feminino , Humanos , Rim , Lipídeos , Lipossomos , Melanoma Experimental/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/patologia , Prolactina/biossíntese , Prolactina/genética , Compostos de Amônio Quaternário , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Transfecção/métodos
7.
Mol Ther ; 15(12): 2094-100, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17726458

RESUMO

Tumor metastases, the most fearsome aspect of cancer, are generally resistant to conventional therapies. Angiogenesis is a crucial aspect of tumor growth and metastatic dissemination. Antiangiogenic therapy, therefore, holds potential as an attractive strategy for inhibiting metastasis development. Human 16K PRL (16K hPRL), a potent inhibitor of angiogenesis, has been demonstrated to prevent tumor growth in two xenograft mouse models, but whether it also affects tumor metastasis is unknown. In this study we will investigate the ability of 16K hPRL to prevent the establishment of metastasis. We demonstrate that 16K hPRL administered via adenovirus-mediated gene transfer, inhibits tumor growth by 86% in a subcutaneous (SC) B16-F10 mouse melanoma model. Computer-assisted image analysis shows that 16K hPRL treatment results in a reduction of tumor-vessel length and width, leading to a 57% reduction of average vessel size. In a pre-established tumor model, moreover, 16K hPRL can significantly delay tumor development. Finally, for the first time, we provide evidence that 16K hPRL considerably reduces the establishment of B16-F10 metastasis in an experimental lung metastasis model. Both the number and size of metastases are reduced by 50% in 16K hPRL-treated mice. These results highlight a potential role for 16K hPRL in anticancer therapy for both primary tumors and metastases.


Assuntos
Adenoviridae/genética , Inibidores da Angiogênese/farmacologia , Divisão Celular/genética , Técnicas de Transferência de Genes , Metástase Neoplásica , Prolactina/farmacologia , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/genética , Animais , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Prolactina/administração & dosagem , Prolactina/genética
8.
Mol Endocrinol ; 21(6): 1422-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17405903

RESUMO

The 16-kDa N-terminal fragment of human prolactin (16K hPRL) is a potent angiostatic factor that inhibits tumor growth in mouse models. Using microarray experiments, we have dissected how the endothelial-cell genome responds to 16K hPRL treatment. We found 216 genes that show regulation by 16K hPRL, of which a large proportion turned out to be associated with the process of immunity. 16K hPRL induces expression of various chemokines and endothelial adhesion molecules. These expressions, under the control of nuclear factor-kappaB, result in an enhanced leukocyte-endothelial cell interaction. Furthermore, analysis of B16-F10 tumor tissues reveals a higher expression of adhesion molecules (intercellular adhesion molecule 1, vascular cell adhesion molecule 1, or E-selectin) in endothelial cells and a significantly higher number of infiltrated leukocytes within the tumor treated with 16K hPRL compared with the untreated ones. In conclusion, this study describes a new antitumor mechanism of 16K hPRL. Because cellular immunity against tumor cells is a crucial step in therapy, the discovery that treatment with 16K hPRL overcomes tumor-induced anergy may become important for therapeutic perspectives.


Assuntos
Proteínas Angiostáticas/farmacologia , Anergia Clonal/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , NF-kappa B/fisiologia , Fragmentos de Peptídeos/farmacologia , Prolactina/farmacologia , Animais , Adesão Celular , Anergia Clonal/genética , Endotélio Vascular/imunologia , Humanos , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , NF-kappa B/genética , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia
9.
Proc Natl Acad Sci U S A ; 103(39): 14319-24, 2006 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-16973751

RESUMO

Angiogenesis is a crucial step in many pathologies, including tumor growth and metastasis. Here, we show that tilted peptides exert antiangiogenic activity. Tilted (or oblique-oriented) peptides are short peptides known to destabilize membranes and lipid cores and characterized by an asymmetric distribution of hydrophobic residues along the axis when helical. We have previously shown that 16-kDa fragments of the human prolactin/growth hormone (PRL/GH) family members are potent angiogenesis inhibitors. Here, we demonstrate that all these fragments possess a 14-aa sequence having the characteristics of a tilted peptide. The tilted peptides of human prolactin and human growth hormone induce endothelial cell apoptosis, inhibit endothelial cell proliferation, and inhibit capillary formation both in vitro and in vivo. These antiangiogenic effects are abolished when the peptides' hydrophobicity gradient is altered by mutation. We further demonstrate that the well known tilted peptides of simian immunodeficiency virus gp32 and Alzheimer's beta-amyloid peptide are also angiogenesis inhibitors. Taken together, these results point to a potential new role for tilted peptides in regulating angiogenesis.


Assuntos
Inibidores da Angiogênese/química , Hormônio do Crescimento/química , Neovascularização Fisiológica/fisiologia , Fragmentos de Peptídeos/química , Prolactina/química , Doença de Alzheimer , Sequência de Aminoácidos , Peptídeos beta-Amiloides/química , Animais , Apoptose/fisiologia , Bovinos , Proliferação de Células , Embrião de Galinha , Células Endoteliais/citologia , Produtos do Gene env/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lipossomos/metabolismo , Fusão de Membrana/fisiologia , Dados de Sequência Molecular , Peso Molecular , Proteínas Mutantes/metabolismo , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Oncogênicas de Retroviridae/química , Proteínas Virais de Fusão/química
10.
Mol Endocrinol ; 19(7): 1932-42, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15746189

RESUMO

The 16-kDa N-terminal fragment of human prolactin (16K hPRL) is a potent antiangiogenic factor that has been shown to prevent tumor growth in a xenograph mouse model. In this paper we first demonstrate that 16K hPRL inhibits serum-induced DNA synthesis in adult bovine aortic endothelial cells. This inhibition is associated with cell cycle arrest at both the G(0)-G(1) and the G(2)-M phase. Western blot analysis revealed that 16K hPRL strongly decreases levels of cyclin D1 and cyclin B1, but not cyclin E. The effect on cyclin D1 is at least partially transcriptional, because treatment with 16K hPRL both reduces the cyclin D1 mRNA level and down-regulates cyclin D1 promoter activity. This regulation may be due to inhibition of the MAPK pathway, but it is independent of the glycogen synthase kinase-3beta pathway. Lastly, 16K hPRL induces the expression of negative cell cycle regulators, the cyclin-dependent kinase inhibitors p21(cip1) and p27(kip1). In summary, 16K hPRL inhibits serum-induced proliferation of endothelial cells through combined effects on positive and negative regulators of cell cycle progression.


Assuntos
Inibidores da Angiogênese/farmacologia , Proteínas de Ciclo Celular/metabolismo , Células Endoteliais/efeitos dos fármacos , Interfase/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Prolactina/farmacologia , Animais , Bovinos , Proteínas de Ciclo Celular/genética , Células Cultivadas , Ciclina B/genética , Ciclina B/metabolismo , Ciclina B1 , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Replicação do DNA/efeitos dos fármacos , Células Endoteliais/metabolismo , Fase G1/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Interfase/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mitose/efeitos dos fármacos , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fase S/efeitos dos fármacos , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
11.
Invest Ophthalmol Vis Sci ; 45(7): 2413-9, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15223825

RESUMO

PURPOSE: To examine the ability and mechanism of the 16 kDa N-terminal fragment of human prolactin (16K hPRL) in the inhibition of abnormal retinal neovascularization. METHODS: The 16K hPRL-encoding sequence was inserted into an adenoviral vector (16K-Ad). Western blot analysis verified the expression of 16K hPRL and inhibition of proliferation, confirming functional activity of the 16K hPRL in virus-infected adult bovine aortic endothelial (ABAE) cells. 16K hPRL inhibited retinal neovascularization in a mouse model of oxygen-induced retinopathy. The ability of recombinant 16K hPRL expressed in E. coli (r16K hPRL) was compared to that of endostatin in inducing apoptosis of cultured human retinal endothelial cells (HREC). RESULTS: 16K was expressed in virus-infected ABAE cells and resulted in a dose-dependent inhibition of cell proliferation. Eyes injected with 16K-Ad showed a reduction in preretinal neovascularization of 82.3 +/- 9.3% (P < 0.00001) when compared to uninjected controls. r16K hPRL was 100 times more potent than endostatin in inducing apoptosis in HRECs. CONCLUSIONS: Intravitreal administration of 16K hPRL inhibited neovascularization in the mouse model of oxygen-induced retinopathy. 16K hPRL stimulated apoptosis in HRECs and inhibited cell proliferation in ABAE cells. These results suggested a potential therapeutic role for 16K hPRL in the treatment of proliferative retinopathies.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Oxigênio/toxicidade , Fragmentos de Peptídeos/uso terapêutico , Prolactina/uso terapêutico , Retina/efeitos dos fármacos , Neovascularização Retiniana/prevenção & controle , Adenoviridae/genética , Inibidores da Angiogênese/genética , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Endostatinas/farmacologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Escherichia coli/metabolismo , Feminino , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/genética , Gravidez , Prolactina/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapêutico , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...