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.
EMBO Mol Med ; 9(12): 1626-1628, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29127101
2.
J Clin Invest ; 126(9): 3495-510, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27548530

RESUMO

The angiopoietin/Tie (ANG/Tie) receptor system controls developmental and tumor angiogenesis, inflammatory vascular remodeling, and vessel leakage. ANG1 is a Tie2 agonist that promotes vascular stabilization in inflammation and sepsis, whereas ANG2 is a context-dependent Tie2 agonist or antagonist. A limited understanding of ANG signaling mechanisms and the orphan receptor Tie1 has hindered development of ANG/Tie-targeted therapeutics. Here, we determined that both ANG1 and ANG2 binding to Tie2 increases Tie1-Tie2 interactions in a ß1 integrin-dependent manner and that Tie1 regulates ANG-induced Tie2 trafficking in endothelial cells. Endothelial Tie1 was essential for the agonist activity of ANG1 and autocrine ANG2. Deletion of endothelial Tie1 in mice reduced Tie2 phosphorylation and downstream Akt activation, increased FOXO1 nuclear localization and transcriptional activation, and prevented ANG1- and ANG2-induced capillary-to-venous remodeling. However, in acute endotoxemia, the Tie1 ectodomain that is responsible for interaction with Tie2 was rapidly cleaved, ANG1 agonist activity was decreased, and autocrine ANG2 agonist activity was lost, which led to suppression of Tie2 signaling. Tie1 cleavage also occurred in patients with hantavirus infection. These results support a model in which Tie1 directly interacts with Tie2 to promote ANG-induced vascular responses under noninflammatory conditions, whereas in inflammation, Tie1 cleavage contributes to loss of ANG2 agonist activity and vascular stability.


Assuntos
Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Inflamação , Receptor de TIE-1/metabolismo , Receptor TIE-2/metabolismo , Remodelação Vascular , Adulto , Idoso , Animais , Estudos de Casos e Controles , Estudos de Coortes , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Endotoxemia/metabolismo , Feminino , Deleção de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Integrina beta1/metabolismo , Lipopolissacarídeos/química , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fosforilação , Sepse , Transdução de Sinais , Adulto Jovem
3.
Oncoimmunology ; 4(6): e1008871, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26155427

RESUMO

Myeloid cells contribute to increased malignancy and poor prognosis in breast cancer. We demonstrate that anti-CSF-1R therapy depletes a cell population sharing characteristics of tumor-associated macrophages (TAMs) and dendritic cells (DCs). Intravital imaging combined with cellular characterization has refined our understanding of anti-CSF-1R therapy on the tumor microenvironment.

4.
Proc Natl Acad Sci U S A ; 111(47): E5086-95, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25385645

RESUMO

Tumor-infiltrating inflammatory cells comprise a major part of the stromal microenvironment and support cancer progression by multiple mechanisms. High numbers of tumor myeloid cells correlate with poor prognosis in breast cancer and are coupled with the angiogenic switch and malignant progression. However, the specific roles and regulation of heterogeneous tumor myeloid populations are incompletely understood. CSF-1 is a major myeloid cell mitogen, and signaling through its receptor CSF-1R is also linked to poor outcomes. To characterize myeloid cell function in tumors, we combined confocal intravital microscopy with depletion of CSF-1R-dependent cells using a neutralizing CSF-1R antibody in the mouse mammary tumor virus long-terminal region-driven polyoma middle T antigen breast cancer model. The depleted cells shared markers of tumor-associated macrophages and dendritic cells (M-DCs), matching the phenotype of tumor dendritic cells that take up antigens and interact with T cells. We defined functional subgroups within the M-DC population by imaging endocytic and matrix metalloproteinase activity. Anti-CSF-1R treatment altered stromal dynamics and impaired both survival of M-DCs and accumulation of new M-DCs, but did not deplete Gr-1(+) neutrophils or block doxorubicin-induced myeloid cell recruitment, and had a minimal effect on lung myeloid cells. Nevertheless, prolonged treatment led to delayed tumor growth, reduced vascularity, and decreased lung metastasis. Because the myeloid infiltrate in metastatic lungs differed significantly from that in mammary tumors, the reduction in metastasis may result from the impact on primary tumors. The combination of functional analysis by intravital imaging with cellular characterization has refined our understanding of the effects of experimental targeted therapies on the tumor microenvironment.


Assuntos
Macrófagos/imunologia , Neoplasias Mamárias Experimentais/patologia , Animais , Divisão Celular , Endocitose , Feminino , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Neutrófilos/imunologia , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Receptor de Fator Estimulador de Colônias de Macrófagos/imunologia , Microambiente Tumoral
5.
J Vis Exp ; (92): 51916, 2014 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-25350573

RESUMO

Myeloid cells are the most abundant immune cells within tumors and have been shown to promote tumor progression. Modern intravital imaging techniques enable the observation of live cellular behavior inside the organ but can be challenging in some types of cancer due to organ and tumor accessibility such as intestine. Direct observation of intestinal tumors has not been previously reported. A surgical procedure described here allows direct observation of myeloid cell dynamics within the intestinal tumors in live mice by using transgenic fluorescent reporter mice and injectable tracers or antibodies. For this purpose, a four-color, multi-region, micro-lensed spinning disk confocal microscope that allows long-term continuous imaging with rapid image acquisition has been used. Apc(Min/+) mice that develop multiple adenomas in the small intestine are crossed with c-fms-EGFP mice to visualize myeloid cells and with ACTB-ECFP mice to visualize intestinal epithelial cells of the crypts. Procedures for labeling different tumor components, such as blood vessels and neutrophils, and the procedure for positioning the tumor for imaging through the serosal surface are also described. Time-lapse movies compiled from several hours of imaging allow the analysis of myeloid cell behavior in situ in the intestinal microenvironment.


Assuntos
Adenoma/patologia , Sistemas Computacionais , Neoplasias Intestinais/patologia , Microscopia Confocal/métodos , Células Mieloides/patologia , Animais , Camundongos , Camundongos Transgênicos
6.
Genes Dev ; 28(14): 1592-603, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25030698

RESUMO

Primitive lymphatic vessels are remodeled into functionally specialized initial and collecting lymphatics during development. Lymphatic endothelial cell (LEC) junctions in initial lymphatics transform from a zipper-like to a button-like pattern during collecting vessel development, but what regulates this process is largely unknown. Angiopoietin 2 (Ang2) deficiency leads to abnormal lymphatic vessels. Here we found that an ANG2-blocking antibody inhibited embryonic lymphangiogenesis, whereas endothelium-specific ANG2 overexpression induced lymphatic hyperplasia. ANG2 inhibition blocked VE-cadherin phosphorylation at tyrosine residue 685 and the concomitant formation of button-like junctions in initial lymphatics. The defective junctions were associated with impaired lymph uptake. In collecting lymphatics, adherens junctions were disrupted, and the vessels leaked upon ANG2 blockade or gene deletion. ANG2 inhibition also suppressed the onset of lymphatic valve formation and subsequent valve maturation. These data identify ANG2 as the first essential regulator of the functionally important interendothelial cell-cell junctions that form during lymphatic development.


Assuntos
Angiopoietina-2/metabolismo , Células Endoteliais/metabolismo , Junções Intercelulares/fisiologia , Angiopoietina-2/antagonistas & inibidores , Angiopoietina-2/genética , Animais , Caderinas/metabolismo , Embrião de Mamíferos , Células Endoteliais/citologia , Deleção de Genes , Linfangiogênese/fisiologia , Tecido Linfoide/embriologia , Tecido Linfoide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação
7.
J Natl Cancer Inst ; 104(6): 461-75, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22343031

RESUMO

BACKGROUND: Angiopoietin-2 (Ang2), a ligand for endothelial TEK (Tie2) tyrosine kinase receptor, is induced in hypoxic endothelial cells of tumors, where it promotes tumor angiogenesis and growth. However, the effects of Ang2 on tumor lymphangiogenesis and metastasis are poorly characterized. METHODS: We addressed the effect of Ang2 on tumor progression and metastasis using systemic Ang2 overexpression in mice carrying tumor xenografts, endothelium-specific overexpression of Ang2 in VEC-tTA/Tet-OS-Ang2 transgenic mice implanted with isogenic tumors, and administration of Ang2-blocking antibodies to tumor-bearing immunodeficient mice. Fisher's exact test was used for analysis of metastasis occurrence, and repeated measures one-way analysis of variance was used for the analysis of primary tumor growth curves. Unpaired t test was used for all other analyses. All statistical tests were two-sided. RESULTS: Adenoviral expression of Ang2 increased lymph node and lung metastasis in tumor xenografts. The metastatic burden in the lungs was increased in transgenic mice in which Ang2 expression was induced specifically in the vascular endothelium (tumor burden per grid, VEC-tTA/Tet-OS-Ang2 mice [n = 5] vs control mice [n = 4]: 45.23 vs 12.26 mm(2), difference = 32.67 mm(2), 95% confidence interval = 31.87 to 34.07, P < .001). Ang2-blocking antibodies reduced lymph node and lung metastasis, as well as tumor lymphangiogenesis, and decreased tumor cell homing to the lungs after intravenous injection. In the lung metastases, Ang2 overexpression decreased endothelial integrity, whereas the Ang2-blocking antibodies improved endothelial cell-cell junctions and basement membrane contacts of metastasis-associated lung capillaries. At the cellular level, the Ang2-blocking antibodies induced the internalization of Ang2-Tie2 receptor complexes from endothelial cell-cell junctions in endothelial-tumor cell cocultures. CONCLUSION: Our results indicate that blocking Ang2 inhibits metastatic dissemination in part by enhancing the integrity of endothelial cell-cell junctions.


Assuntos
Inibidores da Angiogênese/farmacologia , Angiopoietina-2/antagonistas & inibidores , Angiopoietina-2/metabolismo , Endotélio Vascular/efeitos dos fármacos , Junções Intercelulares/efeitos dos fármacos , Junções Intercelulares/metabolismo , Neoplasias Pulmonares/irrigação sanguínea , Neovascularização Patológica/metabolismo , Neovascularização Patológica/prevenção & controle , Animais , Anticorpos Bloqueadores/metabolismo , Anticorpos Bloqueadores/farmacologia , Hipóxia Celular , Técnicas de Cocultura , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Endotélio Vascular/patologia , Feminino , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Junções Intercelulares/patologia , Neoplasias Pulmonares/secundário , Linfangiogênese , Metástase Linfática , Melanoma/irrigação sanguínea , Melanoma/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Neovascularização Patológica/tratamento farmacológico , Receptor TIE-2/metabolismo , Vasos Retinianos/efeitos dos fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Transplante Heterólogo , Regulação para Cima
8.
J Neurosci ; 31(44): 15894-903, 2011 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-22049432

RESUMO

The infiltration of monocytes into the lesioned site is a key event in the inflammatory response after spinal cord injury (SCI). We hypothesized that the molecular events governing the infiltration of monocytes into the injured cord involve cooperativity between the upregulation of the chemoattractant stromal cell-derived factor-1 (SDF-1)/CXCL12 in the injured cord and matrix metalloproteinase-9 (MMP-9/gelatinase B), expressed by infiltrating monocytes. SDF-1 and its receptor CXCR4 mRNAs were upregulated in the injured cord, while macrophages immunoexpressed CXCR4. When mice, transplanted with bone marrow cells from green fluorescent protein (GFP) transgenic mice, were subjected to SCI, GFP+ monocytes infiltrated the cord and displayed gelatinolytic activity. In vitro studies confirmed that SDF-1α, acting through CXCR4, expressed on bone marrow-derived macrophages, upregulated MMP-9 and stimulated MMP-9-dependent transmigration across endothelial cell monolayers by 2.6-fold. There was a reduction in F4/80+ macrophages in spinal cord-injured MMP-9 knock-out mice (by 36%) or wild-type mice, treated with the broad-spectrum MMP inhibitor GM6001 (by 30%). Mice were adoptively transferred with myeloid cells and treated with the MMP-9/-2 inhibitor SB-3CT, the CXCR4 antagonist AMD3100, or a combination of both drugs. While either drug resulted in a 28-30% reduction of infiltrated myeloid cells, the combined treatment resulted in a 45% reduction, suggesting that SDF-1 and MMP-9 function independently to promote the trafficking of myeloid cells into the injured cord. Collectively, these observations suggest a synergistic partnership between MMP-9 and SDF-1 in facilitating transmigration of monocytes into the injured spinal cord.


Assuntos
Movimento Celular/fisiologia , Quimiocina CXCL12/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Monócitos/fisiologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Animais , Benzilaminas , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Quimiocina CXCL12/genética , Ciclamos , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Compostos Heterocíclicos/farmacologia , Compostos Heterocíclicos com 1 Anel/farmacologia , Macrófagos , Metaloproteinase 9 da Matriz/deficiência , Camundongos , Camundongos Transgênicos , Monócitos/efeitos dos fármacos , RNA Mensageiro/metabolismo , Traumatismos da Medula Espinal/terapia , Sulfonas/farmacologia , Fatores de Tempo
9.
Curr Opin Genet Dev ; 20(1): 72-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19942428

RESUMO

Tumor stroma, consisting of the extracellular matrix and multiple cell types such as immune cells, fibroblasts and vascular cells, contributes to the malignancy of solid tumors by a variety of mechanisms. Intravital imaging by different microscopy techniques, especially by confocal and multi-photon microscopy, has proven to be a powerful method for analyzing the cell-cell and cell-matrix interactions in the dynamic tumor microenvironments. Intravital imaging has fostered the acquisition of data on parameters such as motility of different cell types in distinct tumor regions or manipulated with defined challenges, kinetics of tumor cell killing by T cells or macrophage-assisted tumor cell extravasation, functionality of the vasculature, protease activity and metabolic state. Achieving the direct observation of intact tumors offered by intravital imaging provides unique insights into tumor biology that will continue to deepen our understanding of the processes leading to malignancy and of the ways they can be targeted.


Assuntos
Comunicação Celular , Microscopia/métodos , Neoplasias/patologia , Células Estromais/patologia , Movimento Celular , Matriz Extracelular/patologia , Fibroblastos/patologia , Humanos , Inflamação/patologia , Metástase Neoplásica , Neoplasias/irrigação sanguínea , Peptídeo Hidrolases/análise
10.
Curr Opin Cell Biol ; 21(2): 154-65, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19230644

RESUMO

Vascular endothelial growth factors and their endothelial tyrosine kinase receptors are central regulators of vasculogenesis, angiogenesis and lymphangiogenesis. VEGF signalling through VEGFR-2 is the major angiogenic pathway, and blockage of VEGF/VEGFR-2 signalling is the first anti-angiogenic strategy for cancer therapy. VEGFR-1 seems to act as a negative regulator of VEGF-mediated angiogenesis during development, and as a stimulator of pathological angiogenesis when activated by its specific ligands PlGF and VEGF-B. PlGF recruits angiogenic macrophages to tumours, and targeting PlGF could therefore be beneficial in cancer. For VEGF-B, with very limited angiogenic potential, a new role has been identified in regulating lipid metabolism in the heart. VEGF-C and VEGF-D induce lymphangiogenesis via VEGFR-3 and have also been shown to be lymphangiogenic in tumours, stimulating metastasis. Mouse models of lymphoedema have established VEGF-C as a promising agent for pro-lymphangiogenic therapy. In addition to lymphangiogenesis, VEGFR-3 has also been shown to be important for angiogenesis, acting together with VEGF/VEGFR-2 and Dll4/Notch signalling to control angiogenic sprouting. Increasing knowledge of the mechanisms regulating (lymph)angiogenesis should enable the development of better agents to combat metastasis and the resistance of tumours towards anti-angiogenic treatment, and of pro-(lymph)angiogenic treatment methods for ischaemic diseases and lymphoedema.


Assuntos
Linfangiogênese/fisiologia , Neovascularização Fisiológica/fisiologia , Isoformas de Proteínas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Angiopoietina-1/metabolismo , Animais , Humanos , Metástase Linfática , Proteínas de Membrana/metabolismo , Miócitos Cardíacos/fisiologia , Receptor TIE-2/metabolismo , Transdução de Sinais/fisiologia
11.
Am J Pathol ; 173(6): 1891-901, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18988807

RESUMO

Vascular endothelial growth factor-C (VEGF-C) is the quintessential lymphangiogenic growth factor that is required for the development of the lymphatic system and is capable of stimulating lymphangiogenesis in adults by activating its receptor, VEGFR-3. Although VEGF-C is a major candidate molecule for the development of prolymphangiogenic therapy for defective lymphatic vessels in lymphedema, the stability of lymph vessels generated by exogenous VEGF-C administration is not currently known. We studied VEGF-C-stimulated lymphangiogenesis in inducible transgenic mouse models in which growth factor expression can be spatially and temporally controlled without side effects, such as inflammation. VEGF-C induction in adult mouse skin for 1 to 2 weeks caused robust lymphatic hyperplasia that persisted for at least 6 months. VEGF-C induced lymphangiogenesis in numerous tissues and organs when expressed in the vascular endothelium in either neonates or adult mice. Very few or no effects were observed in either blood vessels or collecting lymph vessels. Additionally, VEGF-C stimulated lymphangiogenesis in embryos after the onset of lymphatic vessel development. Strikingly, a strong angiogenic effect was observed after VEGF-C induction in vascular endothelium at any point before embryonic day 16.5. Our results indicate that blood vessels can undergo VEGF-C-induced angiogenesis even after down-regulation of VEGFR-3 in embryos; however, transient VEGF-C expression in adults can induce long-lasting lymphatic hyperplasia with no obvious side effects on the blood vasculature.


Assuntos
Embrião de Mamíferos , Hiperplasia/patologia , Linfangiogênese/fisiologia , Vasos Linfáticos , Transgenes , Fator C de Crescimento do Endotélio Vascular/metabolismo , Animais , Biomarcadores/metabolismo , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Glicoproteínas/metabolismo , Humanos , Hiperplasia/metabolismo , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Gravidez , Tirosina/metabolismo , Fator C de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
Blood ; 105(12): 4642-8, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15746084

RESUMO

Angiopoietin 1 (Ang1), a ligand for the receptor tyrosine kinase Tie2, regulates the formation and stabilization of the blood vessel network during embryogenesis. In adults, Ang1 is associated with blood vessel stabilization and recruitment of perivascular cells, whereas Ang2 acts to counter these actions. Recent results from gene-targeted mice have shown that Ang2 is also essential for the proper patterning of lymphatic vessels and that Ang1 can be substituted for this function. In order to characterize the effects of the angiopoietins on lymphatic vessels, we employed viral vectors for overexpression of Ang1 in adult mouse tissues. We found that Ang1 activated lymphatic vessel endothelial proliferation, vessel enlargement, and generation of long endothelial cell filopodia that eventually fused, leading to new sprouts and vessel development. Cutaneous lymphatic hyperplasia was also detected in transgenic mice expressing Ang1 in the basal epidermal cells. Tie2 was expressed in the lymphatic endothelial cells and Ang1 stimulation of these cells resulted in up-regulation of vascular endothelial growth factor receptor 3 (VEGFR-3). Furthermore, a soluble form of VEGFR-3 inhibited the observed lymphatic sprouting. Our results reinforce the concept that Ang1 therapy may be useful in settings of tissue edema.


Assuntos
Angiopoietina-1/fisiologia , Endotélio Vascular/citologia , Hiperplasia/patologia , Sistema Linfático/fisiologia , Neovascularização Fisiológica , Adenoviridae/genética , Animais , Northern Blotting , Proliferação de Células , Células Cultivadas , Clonagem Molecular , Derme/metabolismo , Edema , Endotélio/citologia , Células Epidérmicas , Vetores Genéticos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Estrutura Terciária de Proteína , Receptor TIE-2/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Regulação para Cima , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
13.
Blood ; 104(10): 3198-204, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15271796

RESUMO

Platelet-derived growth factor-D (PDGF-D) is a recently characterized member of the PDGF family with unknown in vivo functions. We investigated the effects of PDGF-D in transgenic mice by expressing it in basal epidermal cells and then analyzed skin histology, interstitial fluid pressure, and wound healing. When compared with control mice, PDGF-D transgenic mice displayed increased numbers of macrophages and elevated interstitial fluid pressure in the dermis. Wound healing in the transgenic mice was characterized by increased cell density and enhanced recruitment of macrophages. Macrophage recruitment was also the characteristic response when PDGF-D was expressed in skeletal muscle or ear by an adeno-associated virus vector. Combined expression of PDGF-D with vascular endothelial growth factor-E (VEGF-E) led to increased pericyte/smooth muscle cell coating of the VEGF-E-induced vessels and inhibition of the vascular leakiness that accompanies VEGF-E-induced angiogenesis. These results show that full-length PDGF-D is activated in tissues and is capable of increasing interstitial fluid pressure and macrophage recruitment and the maturation of blood vessels in angiogenic processes.


Assuntos
Linfocinas/genética , Linfocinas/metabolismo , Macrófagos/fisiologia , Neovascularização Fisiológica/fisiologia , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Vasos Sanguíneos/fisiologia , Movimento Celular/imunologia , Derme/fisiologia , Líquido Extracelular/fisiologia , Humanos , Macrófagos/citologia , Camundongos , Camundongos Transgênicos , Músculo Esquelético/fisiologia , Pressão , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Virais/genética , Cicatrização
14.
FASEB J ; 17(14): 2006-13, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14597670

RESUMO

Vascular endothelial cells are characterized by a high degree of functional and phenotypic plasticity, which is controlled both by their pericellular microenvironment and their intracellular gene expression programs. To gain further insight into the mechanisms regulating the endothelial cell phenotype, we have compared the responses of lymphatic endothelial cells (LECs) and blood vascular endothelial cells (BECs) to vascular endothelial growth factors (VEGFs). VEGFR-3-specific signals are sufficient for LEC but not BEC proliferation, as shown by the ability of the specific ligand VEGF-C156S to stimulate cell cycle entry only in LECs. On the other hand, we found that VEGFR-3 stimulation did not induce LEC cell shape changes typical of VEGFR-2-stimulated LECs, indicating receptor-specific differences in the cytoskeletal responses. Genes induced via VEGFR-2 also differed between BECs and LECs: angiopoietin-2 (Ang-2) was induced via VEGFR-2 in BECs and LECs, but the smooth muscle cell (SMC) chemoattractant BMP-2 was induced only in BECs. Both BECs and LECs were able to promote SMC chemotaxis, but contact with SMCs led to down-regulation of VEGFR-3 expression in BECs in a 3-dimensional coculture system. This was consistent with the finding that VEGFR-3 is down-regulated in vivo at sites of endothelial cell-pericyte/smooth muscle cell contacts. Collectively, these data show intrinsic cell-specific differences of BEC and LEC responses to VEGFs and identify a pericellular regulatory mechanism for VEGFR-3 down-regulation in endothelial cells.


Assuntos
Endotélio/fisiologia , Sistema Linfático/citologia , Fator de Crescimento Transformador beta , Angiopoietina-2/metabolismo , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Comunicação Celular , Divisão Celular , Movimento Celular , Células Cultivadas , Endotélio/citologia , Endotélio/efeitos dos fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Regulação da Expressão Gênica , Humanos , Músculo Liso/fisiologia , Fenótipo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Thromb Haemost ; 90(2): 167-84, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12888864

RESUMO

The lymphatic vasculature is essential for the maintenance of normal fluid balance and for the immune responses, but it is also involved in a variety of diseases. Hypoplasia or dysfunction of the lymphatic vessels can lead to lymphedema, whereas hyperplasia or abnormal growth of these vessels are associated with lymphangiomas and lymphangiosarcomas. Lymphatic vessels are also involved in lymph node and systemic metastasis of cancer cells. Recent novel findings on the molecular mechanisms involved in lymphatic vessel development and regulation allow the modulation of the lymphangiogenic process and specific targeting of the lymphatic endothelium. Recent results show that the homeodomain transcription factor Prox-1 is an important lymphatic endothelial cell (LEC) fate-determining factor which can induce LEC-specific gene transcription even in blood vascular endothelial cells (BECs). This suggests that the distinct phenotypes of cells in the adult vascular endothelium are plastic and sensitive to transcriptional reprogramming, which might be useful for future therapeutic applications involving endothelial cells. Vascular endothelial growth factor-C (VEGF-C) and VEGF-D are peptide growth factors capable of inducing the growth of new lymphatic vessels in vivo in a process called lymphangiogenesis. They belong to the larger family which also includes VEGF, placenta growth factor (PlGF) and VEGF-B, VEGF-C and VEGF-D are ligands for the endothelial cell specific tyrosine kinase receptors VEGFR-2 and VEGFR-3. In adult human as well as mouse tissues VEGFR-3 is expressed predominantly in lymphatic endothelial cells which line the inner surface of lymphatic vessels. While VEGFR-2 is thought to be the main mediator of angiogenesis, VEGFR-3 signaling is crucial for the development of the lymphatic vessels. Heterozygous inactivation of the VEGFR-3 tyrosine kinase leads to primary lymphedema due to defective lymphatic drainage in the limbs. Other factors that seem to be involved in lymphangiogenesis include the Tie/angiopoietin system, neuropilin-2 and integrin alpha 9. VEGF-C induces lymphatic vessel growth, but high levels of VEGF-C also resulted in blood vessel leakiness and growth. The VEGFR-3-specific mutant form of VEGF-C called VEGF-C156S lacks blood vascular side effects but is sufficient for therapeutic lymphangiogenesis in a mouse model of lymphedema. As VEGF-C156S is a specific lymphatic endothelial growth factor in the skin, it provides an attractive molecule for pro-lymphangiogenic therapy.


Assuntos
Substâncias de Crescimento/fisiologia , Linfangiogênese/fisiologia , Doenças Linfáticas/terapia , Receptores de Superfície Celular/fisiologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...