Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Am J Pathol ; 189(4): 924-939, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30878136

RESUMEN

Obese adipose tissue expansion is an inflammatory process that results in dysregulated lipolysis, increased circulating lipids, ectopic lipid deposition, and systemic insulin resistance. Lymphatic vessels provide a route of fluid, macromolecule, and immune cell clearance, and lymphangiogenesis increases this capability. Indeed, inflammation-associated lymphangiogenesis is critical in resolving acute and chronic inflammation, but it is largely absent in obese adipose tissue. Enhancing adipose tissue lymphangiogenesis could, therefore, improve metabolism in obesity. To test this hypothesis, transgenic mice with doxycycline-inducible expression of murine vascular endothelial growth factor (VEGF)-D under a tightly controlled Tet-On promoter were crossed with adipocyte-specific adiponectin-reverse tetracycline-dependent transactivator mice (Adipo-VD) to stimulate adipose tissue-specific lymphangiogenesis during 16-week high-fat diet-induced obesity. Adipose VEGF-D overexpression induced de novo lymphangiogenesis in murine adipose tissue, and obese Adipo-VD mice exhibited enhanced glucose clearance, lower insulin levels, and reduced liver triglycerides. On ß-3 adrenergic stimulation, Adipo-VD mice exhibited more rapid and increased glycerol flux from adipose tissue, suggesting that the lymphatics are a potential route of glycerol clearance. Resident macrophage crown-like structures were scarce and total F4/80+ macrophages were reduced in obese Adipo-VD s.c. adipose tissue with evidence of increased immune trafficking from the tissue. Augmenting VEGF-D signaling and lymphangiogenesis specifically in adipose tissue, therefore, reduces obesity-associated immune accumulation and improves metabolic responsiveness.


Asunto(s)
Adiponectina/fisiología , Tejido Adiposo/fisiología , Linfangiogénesis , Vasos Linfáticos/fisiología , Obesidad/metabolismo , Factor D de Crecimiento Endotelial Vascular/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Resistencia a la Insulina , Lipólisis , Masculino , Ratones , Ratones Obesos , Ratones Transgénicos , Obesidad/etiología , Obesidad/patología
2.
Development ; 144(3): 507-518, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28087639

RESUMEN

Vascular endothelial growth factors (VEGFs) control angiogenesis and lymphangiogenesis during development and in pathological conditions. In the zebrafish trunk, Vegfa controls the formation of intersegmental arteries by primary angiogenesis and Vegfc is essential for secondary angiogenesis, giving rise to veins and lymphatics. Vegfd has been largely thought of as dispensable for vascular development in vertebrates. Here, we generated a zebrafish vegfd mutant by genome editing. vegfd mutants display significant defects in facial lymphangiogenesis independent of vegfc function. Strikingly, we find that vegfc and vegfd cooperatively control lymphangiogenesis throughout the embryo, including during the formation of the trunk lymphatic vasculature. Interestingly, we find that vegfd and vegfc also redundantly drive artery hyperbranching phenotypes observed upon depletion of Flt1 or Dll4. Epistasis and biochemical binding assays suggest that, during primary angiogenesis, Vegfd influences these phenotypes through Kdr (Vegfr2) rather than Flt4 (Vegfr3). These data demonstrate that, rather than being dispensable during development, Vegfd plays context-specific indispensable and also compensatory roles during both blood vessel angiogenesis and lymphangiogenesis.


Asunto(s)
Linfangiogénesis/fisiología , Neovascularización Fisiológica/fisiología , Factor D de Crecimiento Endotelial Vascular/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Pez Cebra/fisiología , Animales , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Linfangiogénesis/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Modelos Biológicos , Mutagénesis , Neovascularización Fisiológica/genética , Eliminación de Secuencia , Transducción de Señal , Regulación hacia Arriba , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/fisiología , Factor D de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
4.
Blood ; 123(7): 1102-12, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24269955

RESUMEN

Vascular endothelial growth factor-D (VEGFD) is a potent pro-lymphangiogenic molecule during tumor growth and is considered a key therapeutic target to modulate metastasis. Despite roles in pathological neo-lymphangiogenesis, the characterization of an endogenous role for VEGFD in vascular development has remained elusive. Here, we used zebrafish to assay for genetic interactions between the Vegf/Vegf-receptor pathway and SoxF transcription factors and identified a specific interaction between Vegfd and Sox18. Double knockdown zebrafish embryos for Sox18/Vegfd and Sox7/Vegfd exhibit defects in arteriovenous differentiation. Supporting this observation, we found that Sox18/Vegfd double but not single knockout mice displayed dramatic vascular development defects. We find that VEGFD-mitogen-activated protein kinase kinase-extracellular signal-regulated kinase signaling modulates SOX18-mediated transcription, functioning at least in part by enhancing nuclear concentration and transcriptional activity in vascular endothelial cells. This work suggests that VEGFD-mediated pathologies include or involve an underlying dysregulation of SOXF-mediated transcriptional networks.


Asunto(s)
Vasos Sanguíneos/embriología , Neovascularización Fisiológica/genética , Factores de Transcripción SOXF/metabolismo , Factor D de Crecimiento Endotelial Vascular/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Embrión de Mamíferos , Embrión no Mamífero , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción SOXF/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
5.
J Dent Res ; 92(12): 1074-80, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24029591

RESUMEN

Lymphatic vessels return extravasated fluid, proteins, and cells back into the circulation and are important in immune cell trafficking. In the gingiva, lymphatic vessels are located in the lamina propria and travel over the external surface of the alveolar bone. The gingival lymphatics are important for fluid drainage, since lack of lymphatics has been shown to increase interstitial fluid pressure and fluid volume. Maintenance of gingival lymphatic vessels requires continuous signaling by the growth factors VEGF-C and -D via their receptor VEGFR-3. The growth factors are expressed in the gingival epithelium and also in immune cells in the lamina propria. VEGF-C seems to be crucial for lymphangiogenesis induced during periodontal disease development. The lymphatic vessels protect against periodontitis in mice, probably by clearing bacteria and bacterial products and by promoting humoral immune responses. Down-regulation of CCL21, a ligand important for dendritic cell migration, has been demonstrated in lymphatics from patients with periodontitis. High enzymatic activity in the gingiva of these patients may also contribute to impaired lymphatic function, due to the loss of structural components in the interstitium influencing lymphatic function. So far, knowledge is limited in this field because of the dearth of studies on the role of lymphatic vessels in periodontal disease.


Asunto(s)
Linfangiogénesis/fisiología , Vasos Linfáticos/fisiopatología , Enfermedades Periodontales/fisiopatología , Animales , Quimiocina CCL21/fisiología , Líquido Extracelular/fisiología , Humanos , Inmunidad Humoral/inmunología , Linfa/fisiología , Factor C de Crecimiento Endotelial Vascular/fisiología , Factor D de Crecimiento Endotelial Vascular/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/fisiología
7.
Clin Exp Metastasis ; 30(6): 819-32, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23591595

RESUMEN

Metastasis to regional lymph nodes is an important and early event in many tumors. Vascular endothelial growth factor-C (VEGF-C), VEGF-D and their receptor VEGFR-3, play a role in tumor spread via the lymphatics, although the timing of their involvement is not understood. In contrast, VEGFR-2, activated by VEGF-A, VEGF-C and VEGF-D, is a mediator of angiogenesis and drives primary tumor growth. We demonstrate the critical role for VEGFR-3, but not VEGFR-2, in the early events of metastasis. In a tumor model exhibiting both VEGF-D-dependent angiogenesis and lymphangiogenesis, an antibody to VEGFR-2 (DC101) was capable of inhibiting angiogenesis (79 % reduction in PECAM + blood vessels) and growth (93 % reduction in tumor volume). However, unlike an anti-VEGFR-3 Mab (mF4-31C1), DC101 was not capable of eliminating either tumor lymphangiogenesis or lymphogenous metastasis (60 % reduction of lymph node metastasis by DC101 vs 95 % by mF4-31C1). Early excision of the primary tumors demonstrated that VEGF-D-mediated tumor spread precedes angiogenesis-induced growth. Small but highly metastatic primary human breast cancers had significantly higher lymphatic vessel density (23.1 vessels/mm(2)) than size-matched (11.7) or larger non-metastatic tumors (12.4) thus supporting the importance of lymphatic vessels, as opposed to angiogenesis-mediated primary tumor growth, for nodal metastasis. These results suggest that lymphangiogenesis via VEGF-D is more critical than angiogenesis for nodal metastasis.


Asunto(s)
Linfangiogénesis/fisiología , Transducción de Señal/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/fisiología , Animales , Línea Celular Tumoral , Femenino , Humanos , Metástasis Linfática , Vasos Linfáticos/patología , Ratones , Ratones SCID , Neovascularización Fisiológica , Factor D de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología
8.
J Invest Dermatol ; 133(8): 2074-84, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23439394

RESUMEN

The lymphatic vasculature is important for skin biology as it maintains dermal fluid homeostasis. However, the molecular determinants of the form and function of the lymphatic vasculature in skin are poorly understood. Here, we explore the role of vascular endothelial growth factor-d (Vegf-d), a lymphangiogenic glycoprotein, in determining the form and function of the dermal lymphatic network, using Vegf-d-deficient mice. Initial lymphatic vessels in adult Vegf-d-deficient mice were significantly smaller than wild-type but collecting lymphatics were unaltered. The uptake/transport of dextran in initial lymphatics of Vegf-d-deficient mice was far less efficient, indicating compromised function of these vessels. The role of Vegf-d in modulating initial lymphatics was further supported by delivery of Vegf-d in skin of wild-type mice, which promoted enlargement of these vessels. Vegf-d-deficient mice were subjected to cutaneous wounding to challenge lymphatic function: the resulting wound epithelium was highly edematous and thicker, reflecting inadequate lymphatic drainage. Unexpectedly, myofibroblasts were more abundant in Vegf-d-deficient wounds leading to faster wound closure, but resorption of granulation tissue was compromised suggesting poorer-quality healing. Our findings demonstrate that Vegf-d deficiency alters the caliber of initial lymphatics in the dermis leading to reduced functional capacity.


Asunto(s)
Dermis/fisiología , Vasos Linfáticos/anomalías , Vasos Linfáticos/fisiología , Factor D de Crecimiento Endotelial Vascular/fisiología , Cicatrización de Heridas/fisiología , Factores de Edad , Animales , Líquidos Corporales/metabolismo , Dermis/irrigación sanguínea , Dermis/lesiones , Femenino , Tejido de Granulación/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Factor D de Crecimiento Endotelial Vascular/deficiencia , Factor D de Crecimiento Endotelial Vascular/genética
9.
J Biol Chem ; 288(12): 8176-8186, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23404505

RESUMEN

VEGF-D is an angiogenic and lymphangiogenic glycoprotein that can be proteolytically processed generating various forms differing in subunit composition due to the presence or absence of N- and C-terminal propeptides. These propeptides flank the central VEGF homology domain, that contains the binding sites for VEGF receptors (VEGFRs), but their biological functions were unclear. Characterization of propeptide function will be important to clarify which forms of VEGF-D are biologically active and therefore clinically relevant. Here we use VEGF-D mutants deficient in either propeptide, and in the capacity to process the remaining propeptide, to monitor the functions of these domains. We report for the first time that VEGF-D binds heparin, and that the C-terminal propeptide significantly enhances this interaction (removal of this propeptide from full-length VEGF-D completely prevents heparin binding). We also show that removal of either the N- or C-terminal propeptide is required for VEGF-D to drive formation of VEGFR-2/VEGFR-3 heterodimers which have recently been shown to positively regulate angiogenic sprouting. The mature form of VEGF-D, lacking both propeptides, can also promote formation of these receptor heterodimers. In a mouse tumor model, removal of only the C-terminal propeptide from full-length VEGF-D was sufficient to enhance angiogenesis and tumor growth. In contrast, removal of both propeptides is required for high rates of lymph node metastasis. The findings reported here show that the propeptides profoundly influence molecular interactions of VEGF-D with VEGF receptors, co-receptors, and heparin, and its effects on tumor biology.


Asunto(s)
Heparina/química , Factor D de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular , Cromatografía de Afinidad , Células Endoteliales/metabolismo , Femenino , Humanos , Linfangiogénesis , Metástasis Linfática , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Neovascularización Patológica/metabolismo , Neuropilinas/metabolismo , Unión Proteica , Multimerización de Proteína , Precursores de Proteínas/química , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Precursores de Proteínas/fisiología , Estructura Terciaria de Proteína , Eliminación de Secuencia , Factor D de Crecimiento Endotelial Vascular/química , Factor D de Crecimiento Endotelial Vascular/genética , Factor D de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/química
10.
Kidney Int ; 83(1): 50-62, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22932121

RESUMEN

Lymphatic remodeling in inflammation has been found in tracheal mycoplasma infection, human kidney transplant, skin inflammation, peritonitis, and corneal inflammation. Here we investigated lymphangiogenesis in fibrotic area in unilateral ureteral obstruction, a model of progressive renal fibrosis, and evaluated the roles of vascular endothelial growth factor (VEGF)-C and -D in the obstructed kidney. Compared to sham-operated mice, the number of LYVE-1-positive lymphatic vessels, the proliferation of LYVE-1-positive lymphatic endothelial cells, along with VEGF-C and -D mRNA expression were all significantly increased following ureteral obstruction. Depletion of macrophages with clodronate decreased lymphangiogenesis in the obstructed kidney. VEGF-C expression was higher in M2- than in M1-polarized macrophages from bone marrow-derived macrophages, and also increased in Raw 264.7 or renal proximal tubule cells by stimulation with TGF-ß1 or TNF-α. VEGF-D reversed the inhibitory effect of TGF-ß1 on VEGF-C-induced migration, capillary-like tube formation, and proliferation of human lymphatic endothelial cells. Additionally, the blockade of VEGF-C and VEGF-D signaling decreased obstruction-induced lymphangiogenesis. Thus, VEGF-C and VEGF-D are associated with lymphangiogenesis in the fibrotic kidney in a mouse model of ureteral obstruction.


Asunto(s)
Linfangiogénesis/fisiología , Transducción de Señal/fisiología , Obstrucción Ureteral/patología , Obstrucción Ureteral/fisiopatología , Factor C de Crecimiento Endotelial Vascular/fisiología , Factor D de Crecimiento Endotelial Vascular/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Ácido Clodrónico/farmacología , Modelos Animales de Enfermedad , Endotelio Linfático/patología , Fibrosis , Riñón/metabolismo , Riñón/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta1/farmacología , Factor de Necrosis Tumoral alfa/farmacología
11.
Med Oncol ; 29(4): 2594-600, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22562155

RESUMEN

Breast cancer usually initially metastases to the sentinel lymph nodes (SLNs). Recent studies have demonstrated that tumor cells induce SLN lymphangiogenesis before metastasis in several malignancies. In addition, tumor-derived VEGF-C or VEGF-D may induce lymphangiogenesis and promote lymph node metastasis. To explore the mechanisms of lymph node metastasis in breast cancer, we investigated whether primary tumors induce SLN lymphangiogenesis before metastasis and determined the function of tumor-derived VEGF-C and VEGF-D in SLN lymphangiogenesis. Expression of VEGF-C and VEGF-D was examined using immunohistochemistry in 63 primary breast tumors. No significant relationships between VEGF-C and VEGF-D (P=0.420), and VEGF-C or VEGF-D expression and clinical parameters (age, tumor size, grade, hormonal receptor status, her-2 status) were observed (P>0.05). Expression of the lymphatic-specific markers VEGFR-3, Prox-1 and LYVE-1 was measured using quantitative real-time RT-PCR in uninvolved SLNs from 63 patients and compared to control lymph nodes from patients with benign breast disease. Expression of Prox-1 and LYVE-1 mRNA was significantly higher in uninvolved SLNs from breast cancer patients than that in control lymph nodes (P<0.01). Interestingly, expression of VEGFR-3, Prox-1 and LYVE-1 was significantly higher in SLNs from patients with high VEGF-C-expressing tumors than low VEGF-C-expressing tumors (P<0.05), but not VEGF-D-high-expressing tumors (P>0.05). This study demonstrates that primary breast tumors induce SLN lymphangiogenesis before metastasis occurs and that tumor-derived VEGF-C, but not VEGF-D, plays an important role in SLN lymphangiogenesis in breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Linfangiogénesis , Biopsia del Ganglio Linfático Centinela , Factor C de Crecimiento Endotelial Vascular/fisiología , Factor D de Crecimiento Endotelial Vascular/fisiología , Adulto , Anciano , Femenino , Proteínas de Homeodominio/genética , Humanos , Metástasis Linfática , Persona de Mediana Edad , Proteínas Supresoras de Tumor/genética , Factor C de Crecimiento Endotelial Vascular/análisis , Factor D de Crecimiento Endotelial Vascular/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Proteínas de Transporte Vesicular/genética
12.
J Neurosci Res ; 90(7): 1367-81, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22535492

RESUMEN

Human embryonic stem cell (hESC)-derived dopaminergic (DA) neurons hold potential for treating Parkinson's disease (PD) through cell replacement therapy. Generation of DA neurons from hESCs has been achieved by coculture with the stromal cell line PA6, a source of stromal cell-derived inducing activity (SDIA). However, the factors produced by stromal cells that result in SDIA are largely undefined. We previously reported that medium conditioned by PA6 cells can generate functional DA neurons from NTera2 human embryonal carcinoma stem cells. Here we show that PA6-conditioned medium can induce DA neuronal differentiation in both NTera2 cells and the hESC I6 cell line. To identify the factor(s) responsible for SDIA, we used large-scale microarray analysis of gene expression combined with mass spectrometric analysis of PA6-conditioned medium (CM). The candidate factors, hepatocyte growth factor (HGF), stromal cell-derived factor-1 α (SDF1α), secreted frizzled-related protein 1 (sFRP1), and vascular endothelial growth factor D (VEGFD) were identified, and their concentrations in PA6 CM were established by immunoaffinity capillary electrophoresis. Upon addition of SDF1α, sFRP1, and VEGFD to the culture medium, we observed an increase in the number of cells expressing tyrosine hydroxylase (a marker for DA neurons) and ßIII-tubulin (a marker for immature neurons) in both the NTera2 and I6 cell lines. These results indicate that SDF1α, sFRP1, and VEGFD are major components of SDIA and suggest the potential use of these defined factors to elicit DA differentiation of pluripotent human stem cells for therapeutic intervention in PD.


Asunto(s)
Neuronas Dopaminérgicas/citología , Factores de Crecimiento Nervioso/biosíntesis , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/fisiología , Células Madre Pluripotentes/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Línea Celular Tumoral , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/fisiología , Neuronas Dopaminérgicas/metabolismo , Células Madre de Carcinoma Embrionario/citología , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas de la Membrana/farmacología , Proteínas de la Membrana/fisiología , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Células del Estroma/metabolismo , Tubulina (Proteína)/biosíntesis , Tirosina 3-Monooxigenasa/biosíntesis , Factor D de Crecimiento Endotelial Vascular/biosíntesis , Factor D de Crecimiento Endotelial Vascular/fisiología
13.
Cancer Cell ; 21(2): 181-95, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22340592

RESUMEN

Lymphatic metastasis is facilitated by lymphangiogenic growth factors VEGF-C and VEGF-D that are secreted by some primary tumors. We identified regulation of PGDH, the key enzyme in prostaglandin catabolism, in endothelial cells of collecting lymphatics, as a key molecular change during VEGF-D-driven tumor spread. The VEGF-D-dependent regulation of the prostaglandin pathway was supported by the finding that collecting lymphatic vessel dilation and subsequent metastasis were affected by nonsteroidal anti-inflammatory drugs (NSAIDs), known inhibitors of prostaglandin synthesis. Our data suggest a control point for cancer metastasis within the collecting lymphatic endothelium, which links VEGF-D/VEGFR-2/VEGFR-3 and the prostaglandin pathways. Collecting lymphatics therefore play an active and important role in metastasis and may provide a therapeutic target to restrict tumor spread.


Asunto(s)
Transformación Celular Neoplásica , Endotelio Linfático/metabolismo , Metástasis Linfática/fisiopatología , Prostaglandinas/metabolismo , Factor D de Crecimiento Endotelial Vascular/fisiología , Animales , Antiinflamatorios/farmacología , Endotelio Linfático/patología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfangiogénesis/efectos de los fármacos , Metástasis Linfática/genética , Sistema Linfático/efectos de los fármacos , Sistema Linfático/patología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factor D de Crecimiento Endotelial Vascular/genética , Factor D de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Cancer Lett ; 314(2): 127-36, 2012 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-22071224

RESUMEN

Lymph node metastasis is a major prognostic factor for patients with gallbladder cancer (GBC), and greater understanding of the molecule mechanism of lymph node metastasis in GBC is needed to improve prognosis. VEGF-D has been implicated in the control of lymphangiogenesis in many carcinomas, but the biological function of VEGF-D in human GBC remains unclear. In this study, we analyzed the role of the VEGF-D in human GBC cells and addressed the functional role of VEGF-D using a xenograft mouse model. We examined the expression of VEGF-D in three human gallbladder cancer cell lines. A lentivirus-based effective VEGF-D siRNA vector was infected into GBC NOZ cells. The effect of VEGF-D siRNA on GBC NOZ cells was investigated by cell proliferation assay and invasion assay. Furthermore, we examined the role of VEGF-D-SiRNA on GBC NOZ cells in the mice of subcutaneous and orthotopic xenograft tumor. Our results are as follows: VEGF-D mRNA and protein were expressed in all three GBC cell lines (GBC-SD, NOZ, and SGC-996). We successfully selected D-3/siRNA as the most effective siRNA to silence VEGF-D expression after four VEGF-D siRNA plasmid transfection in NOZ cells. VEGF-D mRNA and protein expression were suppressed by lentivirus-mediated D-3/siRNA. D-3-RNAi-LV inhibited NOZ cells proliferation and invasion ability in vitro. D-3-RNAi-LV inhibited tumor growth and lymphangiogenesis in the NOZ cell subcutaneous xenograft model. D-3-RNAi-LV inhibited lymphangiogenesis and lymphatic metastasis in the NOZ cell orthotopic xenograft model. Furthermore, D-3-RNAi-LV inhibited tumor ascites and hepatic invasion in the NOZ cell orthotopic xenograft model. In conclusion, VEGF-D is involved and plays an important role in GBC progression, suggesting that VEGF-D may be a potential molecular target in the treatment of GBC.


Asunto(s)
Neoplasias de la Vesícula Biliar/patología , Linfangiogénesis , Factor D de Crecimiento Endotelial Vascular/fisiología , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Metástasis Linfática , Ratones , Invasividad Neoplásica , ARN Mensajero/análisis , ARN Interferente Pequeño/genética , Factor D de Crecimiento Endotelial Vascular/análisis , Factor D de Crecimiento Endotelial Vascular/genética
15.
Neuron ; 71(1): 117-30, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-21745642

RESUMEN

The role of neuronal dendrites is to receive and process synaptic inputs. The geometry of the dendritic arbor can undergo neuronal activity-dependent changes that may impact the cognitive abilities of the organism. Here we show that vascular endothelial growth factor D (VEGFD), commonly known as an angiogenic mitogen, controls the total length and complexity of dendrites both in cultured hippocampal neurons and in the adult mouse hippocampus. VEGFD expression is dependent upon basal neuronal activity and requires nuclear calcium-calmodulin-dependent protein kinase IV (CaMKIV) signaling. Suppression of VEGFD expression in the mouse hippocampus by RNA interference causes memory impairments. Thus, nuclear calcium-VEGFD signaling mediates the effect of neuronal activity on the maintenance of dendritic arbors in the adult hippocampus and is required for cognitive functioning. These results suggest that caution be employed in the clinical use of blockers of VEGFD signaling for antiangiogenic cancer therapy.


Asunto(s)
Calcio/fisiología , Dendritas/fisiología , Hipocampo/fisiología , Memoria/fisiología , Neuronas/fisiología , Transducción de Señal/fisiología , Factor D de Crecimiento Endotelial Vascular/fisiología , Animales , Animales Recién Nacidos , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/fisiología , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Espinas Dendríticas/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Hipocampo/citología , Hipocampo/metabolismo , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Receptores AMPA/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/fisiología , Factor D de Crecimiento Endotelial Vascular/metabolismo
16.
FASEB J ; 25(8): 2615-25, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21515745

RESUMEN

VEGF-D is a mitogen for endothelial cells that promotes tumor growth and metastatic spread in animal models, and expression of which correlates with lymph node metastasis in some human cancers. It is secreted from the cell as a full-length form with propeptides flanking a central region containing binding sites for VEGFR-2 and VEGFR-3, receptors that signal for angiogenesis and lymphangiogenesis. The propeptides can be cleaved from VEGF-D, enhancing affinity for VEGFR-2 and VEGFR-3 in vitro; however, the importance of this processing in cancer is unclear. To explore the necessity of processing for the effects of VEGF-D in cancer, we use a mutant full-length form that cannot be processed, and show that, in contrast to full-length VEGF-D that is processed, this mutant does not promote tumor growth and lymph node metastasis in a mouse tumor model. Processing of VEGF-D is required for tumor angiogenesis, lymphangiogenesis, and recruitment of tumor-associated macrophages. These observations may be explained by the requirement of processing for VEGF-D to bind neuropilin receptors and activate VEGFR-2. Our results indicate that proteolytic processing is necessary for VEGF-D to promote the growth and spread of cancer, and suggest that enzymes catalyzing this processing could be targets for antimetastatic therapeutics.


Asunto(s)
Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/fisiopatología , Factor D de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Linfangiogénesis/fisiología , Macrófagos/patología , Macrófagos/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Neoplasias Experimentales/fisiopatología , Neovascularización Patológica/fisiopatología , Neuropilinas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trasplante Heterólogo , Factor D de Crecimiento Endotelial Vascular/genética , Factor D de Crecimiento Endotelial Vascular/fisiología
17.
Anticancer Res ; 30(6): 2361-6, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20651394

RESUMEN

BACKGROUND/AIM: This study was designed to investigate the clinical significance of lymphangiogenic vascular endothelial growth factors C and D, and chemokine receptor CCR7 in the lymphatic spread of gastric cancer. PATIENTS AND METHODS: The expressions of VEGF-C and -D, and CCR7 were examined in 82 gastric tumors showing a discrepancy between the degree of lymphatic invasion (Ly) and the status of lymph node metastasis (N) (Ly+N-: 72, and Ly-N+: 10 patients). RESULTS: High expression of VEGF-C and -D, and CCR7 was present in 88%, 63% and 67% of cases, respectively. The VEGF-C expression was significantly higher in Ly+N- than Ly-N+ (p<0.05), but VEGF-D and CCR7 were not. CCR7 expression was a prognostic factor in the Ly+N- subgroup (p<0.05), but VEGF-C and -D were not. CONCLUSION: VEGF-C and -D and CCR7 may play critical roles in lymphatic invasion in primary tumors. CCR7 expression should provide prognostic information in node-negative gastric cancer patients showing lymphatic invasion.


Asunto(s)
Receptores CCR7/análisis , Neoplasias Gástricas/química , Factor C de Crecimiento Endotelial Vascular/análisis , Factor D de Crecimiento Endotelial Vascular/análisis , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Inmunohistoquímica , Metástasis Linfática , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Pronóstico , Receptores CCR7/fisiología , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Factor C de Crecimiento Endotelial Vascular/fisiología , Factor D de Crecimiento Endotelial Vascular/fisiología
18.
Cancer Sci ; 101(10): 2121-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20626397

RESUMEN

Vascular endothelial growth factor (VEGF)-D induces lymphangiogenesis by activating VEGF receptor (VEGFR)-3, which is expressed mainly by lymphatic endothelial cells. VEGFR-3 has also been detected in several types of malignant cells, but the significance of VEGFR-3 expression by malignant cells remains unclear. We examined the expression and function of VEGF-D/VEGFR-3 in human gastric carcinoma cells. Expression of VEGF-D and VEGFR-3 was analyzed in three human gastric carcinoma cell lines and 29 surgical specimens. cDNA microarray analysis was used to examine the effect of VEGF-D on the expression of genes associated with disease progression in VEGFR-3-expressing KKLS cells. VEGF-D-transfected cells and control cells were transplanted into the gastric wall of nude mice. In 10 of the 29 (34%) gastric carcinoma specimens and two of the three cell lines, cancer cells expressed both VEGF-D and VEGFR-3. In vitro treatment of KKLS cells with exogenous VEGF-D increased expression of cyclin D1 and Bcl-2 and stimulated cell proliferation. VEGF-D transfection into KKLS cells resulted in stimulation of angiogenesis, lymphangiogenesis, and cell proliferation, and in inhibition of apoptosis. VEGF-D may participate in the progression of human gastric carcinoma by acting via autocrine and paracrine mechanisms.


Asunto(s)
Comunicación Autocrina , Neoplasias Gástricas/patología , Factor D de Crecimiento Endotelial Vascular/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/análisis , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor D de Crecimiento Endotelial Vascular/análisis , Factor D de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
19.
Eur J Clin Invest ; 40(5): 422-32, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20534064

RESUMEN

BACKGROUND: Arterial occlusive disease is often associated with diabetes mellitus and hypercholesterolaemia which may reduce angiogenic potential of several growth factors. Accordingly, the usefulness of therapeutic angiogenesis in the presence of diabetes and hypercholesterolaemia has remained unclear. We evaluated angiogenic effects of the mature form of vascular endothelial growth factor-D (VEGF-D(deltaNdeltaC)) in skeletal muscles in the presence of severe diabetes and hypercholesterolaemia. METHODS: Intra muscular injections of adenoviruses encoding human VEGF-D(deltaNdeltaC) (AdVEGF-D(deltaNdeltaC)) were given in the hind limbs of a group of diabetic hypercholesterolaemic rabbits and adenoviruses encoding LacZ (AdLacZ) were used as a control. All animals were killed 6 days after the gene transfer. RESULTS: Capillary count, capillary area, capillary permeability and perfusion were significantly higher in the AdVEGF-D(deltaNdeltaC) transduced muscles compared with the AdLacZ controls. Expressions of endothelial nitric oxide synthase (eNOS) and VEGF receptor(R)-2 were also significantly increased in the VEGF-D(deltaNdeltaC) transduced muscles, along with an increased expression of angiopoietins (Angs) and neuropilin-2 (NP-2). Furthermore, VEGF-D(deltaNdeltaC) gene transfer to the skeletal muscles increased localized recruitment of cells with endothelial progenitor-like characteristics. CONCLUSIONS: VEGF-D(deltaNdeltaC) gene transfer can induce efficient angiogenesis in the presence of severe diabetes and hypercholesterolaemia by upregulating eNOS and VEGFR-2 expression. VEGF-D(deltaNdeltaC) appears to be a promising agent for inducing therapeutic angiogenesis even in cases with severe diabetes and hypercholesterolaemia.


Asunto(s)
Glucemia/metabolismo , Lípidos/sangre , Músculo Esquelético/efectos de los fármacos , Factor D de Crecimiento Endotelial Vascular/fisiología , Animales , Permeabilidad Capilar/fisiología , Diabetes Mellitus , Técnicas de Transferencia de Gen , Hipercolesterolemia/inducido químicamente , Operón Lac , Músculo Esquelético/fisiología , Conejos
20.
Br J Dermatol ; 163(4): 776-80, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20491763

RESUMEN

BACKGROUND: Vascular abnormalities are one of the primary pathological components of systemic sclerosis (SSc). However, it has not been determined if there are also abnormalities in the formation of lymphatic vessels in SSc. OBJECTIVE: To evaluate lymphangiogenic activity in SSc skin. METHODS: The numbers of D2-40-positive lymphatic vessels in skin specimens from healthy control subjects and patients with SSc were counted and compared. Quantitative real-time polymerase chain reaction (PCR) was performed to determine mRNA levels of vascular endothelial growth factor (VEGF)-D and Flt-4 (fms-related tyrosine kinase 4, VEGFR-3, one of the receptors for VEGF-D) in the skin. Serum VEGF-D levels were measured with specific enzyme-linked immunosorbent assays. RESULTSZ: The number of lymphatic vessels in patients with SSc was significantly decreased compared with healthy control subjects. Mean relative transcript levels of FIGF (VEGF-D) and FLT4 (Flt-4) in skin tissue from patients with SSc were significantly increased compared with healthy control subjects. By the analysis of the association between serum VEGF-D levels and the clinical or laboratory features, we found that patients with SSc with higher serum VEGF-D levels more frequently have skin ulcers than those with normal VEGF-D levels. CONCLUSIONS: A systemic increase of VEGF-D, as well as local overexpression of FIGF and FLT4, may be the cause of disturbed lymphangiogenesis in SSc skin and play a role in the pathogenesis of SSc. We showed the possibility that regulation of VEGF-D/Flt-4 signalling could lead to new treatment of skin ulcers in SSc by controlling the formation of lymphatic vessels.


Asunto(s)
Linfangiogénesis/fisiología , Esclerodermia Sistémica/fisiopatología , Piel/metabolismo , Factor D de Crecimiento Endotelial Vascular/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/fisiología , Femenino , Expresión Génica/fisiología , Humanos , Vasos Linfáticos/patología , Masculino , Persona de Mediana Edad , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Transducción de Señal/fisiología , Piel/patología , Factor D de Crecimiento Endotelial Vascular/biosíntesis , Factor D de Crecimiento Endotelial Vascular/sangre , Receptor 3 de Factores de Crecimiento Endotelial Vascular/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...