Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cancer Res Commun ; 2(11): 1504-1519, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36970050

RESUMO

The receptor tyrosine kinase VEGFR-3 plays a crucial role in cancer-induced angiogenesis and lymphangiogenesis, promoting tumor development and metastasis. Here, we report the novel VEGFR-3 inhibitor EVT801 that presents a more selective and less toxic profile than two major inhibitors of VEGFRs (i.e., sorafenib and pazopanib). As monotherapy, EVT801 showed a potent antitumor effect in VEGFR-3-positive tumors, and in tumors with VEGFR-3-positive microenvironments. EVT801 suppressed VEGF-C-induced human endothelial cell proliferation in vitro and tumor (lymph)angiogenesis in different tumor mouse models. In addition to reduced tumor growth, EVT801 decreased tumor hypoxia, favored sustained tumor blood vessel homogenization (i.e., leaving fewer and overall larger vessels), and reduced important immunosuppressive cytokines (CCL4, CCL5) and myeloid-derived suppressor cells (MDSC) in circulation. Furthermore, in carcinoma mouse models, the combination of EVT801 with immune checkpoint therapy (ICT) yielded superior outcomes to either single treatment. Moreover, tumor growth inhibition was inversely correlated with levels of CCL4, CCL5, and MDSCs after treatment with EVT801, either alone or combined with ICT. Taken together, EVT801 represents a promising anti(lymph)angiogenic drug for improving ICT response rates in patients with VEGFR-3 positive tumors. Significance: The VEGFR-3 inhibitor EVT801 demonstrates superior selectivity and toxicity profile than other VEGFR-3 tyrosine kinase inhibitors. EVT801 showed potent antitumor effects in VEGFR-3-positive tumors, and tumors with VEGFR-3-positive microenvironments through blood vessel homogenization, and reduction of tumor hypoxia and limited immunosuppression. EVT801 increases immune checkpoint inhibitors' antitumor effects.


Assuntos
Neoplasias , Receptor 3 de Fatores de Crescimento do Endotélio Vascular , Humanos , Camundongos , Animais , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Neoplasias/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Imunoterapia , Microambiente Tumoral
2.
J Biol Chem ; 290(6): 3405-17, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25384978

RESUMO

The formation of new vessels in the tumor, termed angiogenesis, is essential for primary tumor growth and facilitates tumor invasion and metastasis. Hypoxia has been described as one trigger of angiogenesis. Indeed, hypoxia, which is characterized by areas of low oxygen levels, is a hallmark of solid tumors arising from an imbalance between oxygen delivery and consumption. Hypoxic conditions have profound effects on the different components of the tumoral environment. For example, hypoxia is able to activate endothelial cells, leading to angiogenesis but also thereby initiating a cascade of reactions involving neutrophils, smooth muscle cells, and fibroblasts. In addition, hypoxia directly regulates the expression of many genes for which the role and the importance in the tumoral environment remain to be completely elucidated. In this study, we used a method to selectively label sialoglycoproteins to identify new membrane and secreted proteins involved in the adaptative process of endothelial cells by mass spectrometry-based proteomics. We used an in vitro assay under hypoxic condition to observe an increase of protein expression or modifications of glycosylation. Then the function of the identified proteins was assessed in a vasculogenesis assay in vivo by using a morpholino strategy in zebrafish. First, our approach was validated by the identification of sialoglycoproteins such as CD105, neuropilin-1, and CLEC14A, which have already been described as playing key roles in angiogenesis. Second, we identified several new proteins regulated by hypoxia and demonstrated for the first time the pivotal role of GLUT-1, TMEM16F, and SDF4 in angiogenesis.


Assuntos
Neovascularização Fisiológica , Processamento de Proteína Pós-Traducional , Sialoglicoproteínas/metabolismo , Adaptação Fisiológica , Animais , Anoctaminas , Antígenos CD/genética , Antígenos CD/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Hipóxia Celular , Endoglina , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilação , Células Endoteliais da Veia Umbilical Humana , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Proteínas de Transferência de Fosfolipídeos/genética , Proteínas de Transferência de Fosfolipídeos/metabolismo , Proteoma/química , Proteoma/metabolismo , Proteômica/métodos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Sialoglicoproteínas/genética , Peixe-Zebra
3.
J Cell Physiol ; 230(1): 43-51, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24760775

RESUMO

Tumor angiogenesis is accompanied by vasculogenesis, which is involved in the differentiation and mobilization of human bone marrow cells. In order to further characterize the role of vasculogenesis in the tumor growth process, the effects of FGF2 on the differentiation of human bone marrow AC133(+) cells (BM-AC133(+)) into vascular precursors were studied in vitro. FGF2, like VEGFA, induced progenitor cell differentiation into cell types with endothelial cell characteristics. SSR128129E, a newly discovered specific FGFR antagonist acting by allosteric interaction with FGFR, abrogated FGF2-induced endothelial cell differentiation, showing that FGFR signaling is essential during this process. To assess the involvement of the FGF/FRGR signaling in vivo, the pre-clinical model of Lewis lung carcinoma (LL2) in mice was used. Subcutaneous injection of LL2 cells into mice induced an increase of circulating EPCs from peripheral blood associated with tumor growth and an increase of intra-tumoral vascular index. Treatment with the FGFR antagonist SSR128129E strongly decreased LL2 tumor growth as well as the intra-tumoral vascular index (41% and 50% decrease vs. vehicle-treated mice respectively, P < 0.01). Interestingly, SSR128129E treatment significantly decreased the number of circulating EPCs from the peripheral blood (53% inhibition vs. vehicle-treated mice, P < 0.01). These results demonstrate for the first time that the blockade of the FGF/FGFR pathway by SSR128129E reduces EPC recruitment during angiogenesis-dependent tumor growth. In this context, circulating EPCs could be a reliable surrogate marker for tumor growth and angiogenic activity.


Assuntos
Carcinoma Pulmonar de Lewis/irrigação sanguínea , Células-Tronco Hematopoéticas/citologia , Indolizinas/farmacologia , Neovascularização Patológica/patologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , ortoaminobenzoatos/farmacologia , Antígeno AC133 , Animais , Antígenos CD/biossíntese , Células da Medula Óssea/metabolismo , Adesão Celular , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular , Células Endoteliais/citologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glicoproteínas/biossíntese , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos , RNA Mensageiro/biossíntese , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
4.
J Lab Autom ; 18(5): 411-5, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23813914

RESUMO

Angiogenesis is the biological process of generating new capillary blood vessels. It is a fundamental component of a number of normal (reproduction and wound healing) and pathological processes (diabetic retinopathy, rheumatoid arthritis, tumor growth, and metastasis). In vitro angiogenesis assays provide a platform for evaluating the effects of pro- or antiangiogenic compounds. One of the most informative assays is the endothelial cells capillary tube formation assay performed on a biological matrix. This assay is based on quantification of the stimulatory and inhibitory effects of various agents, which is estimated through the measurement of the pseudo-tubules network length. This standard measurement is usually carried out manually by trained operators but requires time, attention, and dedication to achieve a reasonable degree of accuracy. Moreover, the screening is operator dependent. In this article, we propose an automated procedure to evaluate the pseudo-tubule network lengths. We propose a series of image analysis procedures developed using a freely available image analysis software library. More than 800 images from 12 experiments were analyzed automatically and manually, and their results were compared to improve and validate the proposed image analysis procedure. The resulting image analysis software is currently running on a dedicated server, with comparable accuracy to manual measurements. Using this new automated procedure, we are able to treat 540 images, or three complete assays per hour.


Assuntos
Técnicas Citológicas/métodos , Células Endoteliais/efeitos dos fármacos , Processamento de Imagem Assistida por Computador/métodos , Neovascularização Fisiológica/efeitos dos fármacos , Humanos , Software
5.
Cancer Cell ; 23(4): 477-88, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23597562

RESUMO

Receptor tyrosine kinases (RTK) are targets for anticancer drug development. To date, only RTK inhibitors that block orthosteric binding of ligands and substrates have been developed. Here, we report the pharmacologic characterization of the chemical SSR128129E (SSR), which inhibits fibroblast growth factor receptor (FGFR) signaling by binding to the extracellular FGFR domain without affecting orthosteric FGF binding. SSR exhibits allosteric properties, including probe dependence, signaling bias, and ceiling effects. Inhibition by SSR is highly conserved throughout the animal kingdom. Oral delivery of SSR inhibits arthritis and tumors that are relatively refractory to anti-vascular endothelial growth factor receptor-2 antibodies. Thus, orally-active extracellularly acting small-molecule modulators of RTKs with allosteric properties can be developed and may offer opportunities to improve anticancer treatment.


Assuntos
Inibidores de Proteínas Quinases/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Regulação Alostérica , Animais , Anticorpos Monoclonais/farmacologia , Artrite Experimental/tratamento farmacológico , Reabsorção Óssea/tratamento farmacológico , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/metabolismo , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Proc Natl Acad Sci U S A ; 106(15): 6152-7, 2009 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-19369214

RESUMO

VEGF-B, a homolog of VEGF discovered a long time ago, has not been considered an important target in antiangiogenic therapy. Instead, it has received little attention from the field. In this study, using different animal models and multiple types of vascular cells, we revealed that although VEGF-B is dispensable for blood vessel growth, it is critical for their survival. Importantly, the survival effect of VEGF-B is not only on vascular endothelial cells, but also on pericytes, smooth muscle cells, and vascular stem/progenitor cells. In vivo, VEGF-B targeting inhibited both choroidal and retinal neovascularization. Mechanistically, we found that the vascular survival effect of VEGF-B is achieved by regulating the expression of many vascular prosurvival genes via both NP-1 and VEGFR-1. Our work thus indicates that the function of VEGF-B in the vascular system is to act as a "survival," rather than an "angiogenic" factor and that VEGF-B inhibition may offer new therapeutic opportunities to treat neovascular diseases.


Assuntos
Neovascularização Patológica , Fator B de Crescimento do Endotélio Vascular/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Genoma , Membro Posterior/irrigação sanguínea , Isquemia/genética , Isquemia/metabolismo , Camundongos , Camundongos Knockout , Ratos , Retina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Regulação para Cima , Fator B de Crescimento do Endotélio Vascular/deficiência , Fator B de Crescimento do Endotélio Vascular/genética
7.
J Reprod Immunol ; 76(1-2): 17-22, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17467060

RESUMO

In this review, we summarize the results of a number of our recent in vitro and in vivo experiments demonstrating that, in addition to the immunoregulatory functions, soluble HLA-G molecules also affect endothelial cell activity. We have found that soluble HLA-G1 (also designated HLA-G5) inhibits endothelial cell proliferation, migration and tubule formation, and this occurred through binding to the CD160 receptor and via an apoptotic pathway. Moreover, we have demonstrated that soluble HLA-G1 blocks in vivo rabbit corneal neoangiogenesis. Although it cannot be excluded that other soluble HLA class I molecules may have similar effects, as soluble forms of HLA-G are being produced by trophoblast cells at the maternal-fetal interface during early gestation, we discuss how such anti-angiogenic properties of soluble HLA-G1 may locally influence uterine vascular remodeling.


Assuntos
Antígenos CD/metabolismo , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Neovascularização Fisiológica , Receptores Imunológicos/metabolismo , Moduladores da Angiogênese/metabolismo , Animais , Células Endoteliais/fisiologia , Feminino , Proteínas Ligadas por GPI , Antígenos HLA-G , Humanos , Placenta/irrigação sanguínea , Placenta/imunologia , Gravidez , Trofoblastos/imunologia , Útero/irrigação sanguínea
8.
Blood ; 108(8): 2608-15, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16809620

RESUMO

HLA-G is a major histocompatibility complex class Ib molecule whose constitutive tissue distribution is restricted mainly to trophoblast cells at the maternal-fetal interface during pregnancy. In this study, we demonstrated the ability of the soluble HLA-G1 (sHLA-G1) isoform to inhibit fibroblast growth factor-2 (FGF2)-induced capillary-like tubule formation. Using a rabbit corneal neovascularization model, we further showed that sHLA-G1 inhibits FGF2-induced angiogenesis in vivo. We also demonstrated that sHLA-G1 induces endothelial cell apoptosis through binding to BY55/CD160, a glycosylphosphatidylinositolanchored receptor expressed by endothelial cells. Furthermore, we showed that the specific CL1-R2 anti-CD160 monoclonal antibody mimics sHLA-G1-mediated inhibition of endothelial cell tube formation and induction of apoptosis. Thus, the engagement of CD160 in endothelial cells may be essential for the inhibition of angiogenesis. sHLA-G1/CD160-mediated antiangiogenic property may participate in the vascular remodeling of maternal spiral arteries during pregnancy, and, given that we found that CD160 is strongly expressed in the vasculature of a murine tumor, it offers an attractive therapeutic target for preventing pathologic neovascularization.


Assuntos
Antígenos CD/metabolismo , Células Endoteliais/efeitos dos fármacos , Antígenos HLA/metabolismo , Antígenos HLA/farmacologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe I/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Receptores Imunológicos/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Apoptose/fisiologia , Células Cultivadas , Córnea/irrigação sanguínea , Córnea/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/imunologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteínas Ligadas por GPI , Antígenos HLA-G , Humanos , Técnicas In Vitro , Camundongos , Dados de Sequência Molecular , Coelhos , Receptores Imunológicos/genética , Solubilidade
9.
Blood ; 108(4): 1243-50, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16621967

RESUMO

Neuropilin 2 (NRP2) is a receptor for the vascular endothelial growth factor (VEGF) and the semaphorin (SEMA) families, 2 unrelated ligand families involved in angiogenesis and neuronal guidance. NRP2 specifically binds VEGF-A and VEGF-C, although the biological relevance of these interactions in human endothelial cells is poorly understood. In this study, we show that both VEGF-A and VEGF-C induce the interaction of NRP2 with VEGFR-2. This interaction correlated with an enhancement of the VEGFR-2 phosphorylation threshold. Overexpression of NRP2 in primary human endothelial cells promoted cell survival induced by VEGF-A and VEGF-C. In contrast, SEMA3F, another ligand for NRP2, was able to inhibit human endothelial cell survival and migration induced by VEGF-A and VEGF-C. Moreover, a siRNA targeting specifically NRP2 was a potent inhibitor of human endothelial cell migration induced by VEGF-A and VEGF-C. Thus, our data indicate that NRP2 acts as a coreceptor that enhances human endothelial cell biological responses induced by VEGF-A and VEGF-C.


Assuntos
Movimento Celular/fisiologia , Células Endoteliais/fisiologia , Neuropilina-2/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Expressão Gênica , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/farmacologia , Neuropilina-2/antagonistas & inibidores , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator C de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/farmacologia
10.
J Clin Invest ; 115(1): 118-27, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15630451

RESUMO

The angiogenic mechanism and therapeutic potential of PDGF-CC, a recently discovered member of the VEGF/PDGF superfamily, remain incompletely characterized. Here we report that PDGF-CC mobilized endothelial progenitor cells in ischemic conditions; induced differentiation of bone marrow cells into ECs; and stimulated migration of ECs. Furthermore, PDGF-CC induced the differentiation of bone marrow cells into smooth muscle cells and stimulated their growth during vessel sprouting. Moreover, delivery of PDGF-CC enhanced postischemic revascularization of the heart and limb. Modulating the activity of PDGF-CC may provide novel opportunities for treating ischemic diseases.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Isquemia/tratamento farmacológico , Isquemia/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Células-Tronco/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Vasos Coronários/citologia , Vasos Coronários/efeitos dos fármacos , Membro Posterior/irrigação sanguínea , Membro Posterior/efeitos dos fármacos , Humanos , Isquemia/induzido quimicamente , Isquemia/metabolismo , Linfocinas , Camundongos , Microcirculação/efeitos dos fármacos , Miocárdio/metabolismo , Miocárdio/patologia , Fosfotirosina/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Células-Tronco/citologia
11.
Biochem Biophys Res Commun ; 324(2): 909-15, 2004 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-15474514

RESUMO

VEGFR-3 is essential for vascular development and maintenance of lymphatic vessel's integrity. Little is known about its cooperative effect with other receptors of the same family. Contrary to VEGFR-2, stimulation of VEGFR-3 by VEGF-C and -D failed to enhance its phosphorylation either in HEK293T or in PAE cells. These ligands were unable to induce angiogenesis of PAEC expressing VEGFR-3 alone. In the presence of VEGFR-2, VEGF-C and -D induced heterodimerization of VEGFR-3 with VEGFR-2. This heterodimerization was associated with enhanced VEGFR-3 phosphorylation and subsequent cellular responses as evidenced by the formation of capillary-like structures in PAE cells and proliferation of primary human endothelial cells expressing both receptors. Taken together, these results show for the first time that VEGFR-3 needs to be associated to VEGFR-2 to induce ligand-dependent cellular responses.


Assuntos
Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/química , Western Blotting , Carbazóis/farmacologia , Linhagem Celular , Proliferação de Células , Células Cultivadas , Dimerização , Células Endoteliais/citologia , Endotélio Vascular/citologia , Humanos , Imunoprecipitação , Ligantes , Neovascularização Patológica , Peptídeos/química , Fosforilação , Ligação Proteica , Proteínas Tirosina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção , Fator C de Crescimento do Endotélio Vascular/química , Fator D de Crescimento do Endotélio Vascular/química
12.
J Cell Physiol ; 200(3): 351-9, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15254962

RESUMO

Tumor growth and metastasis require the generation of new blood vessels, a process known as neo-angiogenesis. Recent studies have indicated that early tumor vascularization is characterized by the differentiation and mobilization of human bone marrow cells. Vascular endothelial growth factor-A (VEGF-A) is one of the growth factors, which enhances their differentiation into endothelial cells, but little is known about the implication of the VEGF-receptor tyrosine kinases and about the implication of the VEGF-R co-receptor, neuropilin-1, in this process. In this context, the identification of the molecular pathways that support the proliferation and differentiation of vascular stem and progenitor cells was investigated in order to define the pharmaceutical targets involved in tissue vascularization associated with this process. For this purpose, an in vitro model of differentiation of human bone marrow AC133+ (BM-AC133+) cells into vascular precursors was used. In this work, we have demonstrated for the first time that the effect of VEGF-A on BM-AC133+ cells relies on an early action of VEGF-A on the expression of its tyrosine kinase receptors followed by an activation of a VEGF-R2/neuropilin-1-dependent signaling pathway. This signaling promotes the differentiation of BM-AC133+ cells into endothelial precursor cells, followed by the proliferation of these differentiated cells. Altogether, these results strongly suggest that VEGF inhibitors, acting at the level of VEGF-R2 and/or neuropilin-1, by inhibiting differentiation and proliferation of these cells, could be potentially active compounds to prevent progenitor cells to be involved in tumor angiogenesis leading to tumor growth.


Assuntos
Células da Medula Óssea/fisiologia , Diferenciação Celular , Células-Tronco Hematopoéticas/fisiologia , Neuropilina-1/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adesão Celular , Divisão Celular , Movimento Celular , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Humanos , Neovascularização Patológica/prevenção & controle , Molécula-1 de Adesão Celular Endotelial a Plaquetas , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...