Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 227
Filtrar
1.
Invest Ophthalmol Vis Sci ; 62(13): 17, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34673901

RESUMO

Purpose: No lymphatic vessels have been identified in the retina. This study investigated whether pathological VEGF-A-overexpressing diabetic retina causes lymphangiogenesis. Methods: Three genetic mouse models of diabetic retinopathy (DR) (Akita [Ins2+/-], Kimba [vegfa+/+], and Akimba [Akita × Kimba] mice) were used. Retinas were examined by fundus photography, fluorescence angiography (FA), and immunostaining to detect lymphangiogenesis or angiogenesis. Lyve1-GFP (Lyve1EGFP/Cre) mice were used to examine Lyve1-expressing cells by immunostaining. Lymphatic-related factors were investigated in mouse retina and vitreous fluid from proliferative diabetic retinopathy (PDR) patients by RT-PCR and ELISA, respectively. Aged Kimba and Akimba mice were used to examine the retinal phenotype at the late phase of VEGF overexpression. Results: FA and immunostaining showed retinal neovascularization in Kimba and Akimba mice but not wild-type and Akita mice. Immunohistochemistry showed that lymphangiogenesis was not present in the retinas of Akita, Kimba, or Akimba mice despite the significant upregulation of lymphatic-related factors (Lyve1, podoplanin, VEGF-A, VEGF-C, VEGF-D, VEGFR2, and VEGFR3) in the retinas of Kimba and Akimba mice by RT-PCR (P < 0.005). Furthermore, lymphangiogenesis was not present in aged Kimba or Akimba mice. Significantly increased numbers of Lyve1-positive cells present in the retinas of Kimba and Akimba mice, especially in the peripheral areas, were CD11b positive, indicating a macrophage population (P < 0.005). VEGF-C in PDR vitreous with vitreous hemorrhage (VH) was higher than in PDR without VH or a macular hole. Conclusions: Retinal VEGF-A overexpression did not cause typical lymphangiogenesis despite upregulated lymphatic-related factors and significant Lyve1-positive macrophage infiltration.


Assuntos
Retinopatia Diabética/genética , Regulação da Expressão Gênica , Linfangiogênese/genética , Vasos Linfáticos/patologia , Retina/metabolismo , Fator C de Crescimento do Endotélio Vascular/genética , Proteínas de Transporte Vesicular/genética , Animais , Diabetes Mellitus Experimental , Retinopatia Diabética/diagnóstico por imagem , Retinopatia Diabética/metabolismo , Angiofluoresceinografia/métodos , Vasos Linfáticos/metabolismo , Camundongos Endogâmicos C57BL , RNA/genética , Retina/patologia , Regulação para Cima , Fator C de Crescimento do Endotélio Vascular/biossíntese , Proteínas de Transporte Vesicular/biossíntese
2.
Cancer Sci ; 112(1): 243-253, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33128283

RESUMO

Pancreatic neuroendocrine tumor (pNET) is a pancreatic neoplasm with neuroendocrine differentiation. pNET in early stage can be treated with surgical resection with long-term survival, whereas the prognosis of pNET with locoregional or distant metastasis is relatively poor. Lymphangiogenesis is essential for tumor metastasis via the lymphatic system and may overhead distant metastasis. c-Myc overexpression is involved in tumorigenesis. The role of c-Myc in lymphangiogenesis is unclear. In this study, we evaluated the mechanism and effect of c-Myc on lymphangiogenesis of pNET via interaction of lymphatic endothelial cells (LECs) and pNET cells. Lymph node metastasis was evaluated in pNET xenograft mice. Potential target agents to inhibit lymph node metastasis were evaluated in an animal model. We found that vascular endothelial growth factor C (VEGFC) expression and secretion was increased in pNET cell lines with c-Myc overexpression. c-Myc transcriptionally upregulates VEGFC expression and the secretion of pNET cells by directly binding to the E-box of the VEGFC promoter and enhances VEGF receptor 3 phosphorylation and the tube formation of LECs. c-Myc overexpression is associated with lymph node metastasis in pNET xenograft mice. Combinational treatment with an mTOR inhibitor and c-Myc inhibitor or VEGFC-neutralizing chimera protein reduced lymph node metastasis in the mice with c-Myc overexpression. The mTOR inhibitor acts on lymphangiogenesis by reducing VEGFC expression in pNET cells and inhibiting the tube formation of LECs. In conclusion, mTOR and c-Myc are important for lymphangiogenesis of pNET and are potential therapeutic targets for prevention and treatment of lymph node metastasis in pNET.


Assuntos
Metástase Linfática/patologia , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator C de Crescimento do Endotélio Vascular/biossíntese , Animais , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos , Humanos , Linfangiogênese/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Regulação para Cima
3.
Biochem Cell Biol ; 98(6): 676-682, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33167678

RESUMO

Although the abundance of long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) in lung cancer has been well researched, the underlying mechanisms behind its effects were unknown. Here we investigated the molecular events regulating PVT1 in lung cancer. The pro-proliferative property of PVT1 was examined using a xenograft tumor model. Transwell chambers were used to analyze the impact of PVT1 expression on cell invasiveness and migration. In vivo metastasis was examined by tail-vein-injection in mice. Direct binding of miR-128 to PVT1 was investigated using a probe pulldown assay. The relative expression levels of miR-128 and PVT1 were quantified by real-time polymerase chain reaction and Western blotting. We show here that when PVT1 is amplified, there is a poor survival prognosis for patients with lung cancer. Elevated levels of PVT1 promoted lung cancer cell proliferation and metastasis, both in vitro and in vivo. Mechanistically, we found that PVT1 competes endogenously with miR-128 in the regulation of vascular endothelial growth factor C (VEGFC) expression, which is significantly associated with an unfavorable prognosis in lung cancer. We identified that copy number amplification significantly contributes to the high level of PVT1 transcripts in lung cancer, which promotes cell proliferation and metastatic behavior via modulating VEGFC expression by endogenous competition with miR-128.


Assuntos
Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/biossíntese , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Fator C de Crescimento do Endotélio Vascular/biossíntese , Células A549 , Animais , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Proteínas de Neoplasias/genética , RNA Longo não Codificante/genética , RNA Neoplásico/genética , Fator C de Crescimento do Endotélio Vascular/genética
4.
Neurosci Lett ; 735: 135197, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32590044

RESUMO

BACKGROUND: Lymphatic vessels (LVs) of meninges and lymphatic drainage in the brain have been investigated previously. Here, we examined the role of continuous theta burst stimulation (CTBS) in the modulation of meningeal LVs. METHODS: To explore the effects of CTBS on meningeal LVs, the diameters of LVs were measured between a real CTBS group and sham CTBS group of wild-type male mice. Vascular endothelial growth factor-C (VEGF-C) expression was subsequently calculated in both groups to account for lymphatic changes after CTBS. Sunitinib was administered by 3-day oral gavage to inhibit the VEGF receptor (VEGFR), and the effects of CTBS were further examined in the following groups: vehicle with real CTBS, vehicle with sham CTBS, sunitinib treatment with real CTBS, and sunitinib treatment with sham CTBS. RESULTS: The lymphatic vessels were augmented, and the level of VEGF-C in meninges increased after CTBS. CTBS dilated meningeal lymphatic vessels were impaired after the VEGF-C/VEGFR3 pathway was blocked. CONCLUSIONS: CTBS can dilate meningeal lymphatic vessels by up-regulating VEGF-C in meninges.


Assuntos
Sistema Glinfático/metabolismo , Vasos Linfáticos/metabolismo , Meninges/metabolismo , Ritmo Teta/fisiologia , Regulação para Cima/fisiologia , Fator C de Crescimento do Endotélio Vascular/biossíntese , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estimulação Magnética Transcraniana/métodos
5.
J Pathol ; 251(3): 323-335, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32418202

RESUMO

The lymphatic system plays a crucial role in the maintenance of tissue fluid homeostasis and the immunological response to inflammation. The effects of lymphatic drainage dysfunction on periodontitis have not been well studied. Here we show that lymphatic vessel endothelial receptor 1 (LYVE1)+ /podoplanin (PDPN)+ lymphatic vessels (LVs) are increased in the periodontal tissues, with accumulation close to the alveolar bone surface, in two murine periodontitis models: rheumatoid arthritis (RA)-associated periodontitis and ligature-induced periodontitis. Further, PDPN+ /alpha-smooth muscle actin (αSMA)- lymphatic capillaries are increased, whereas PDPN+ /αSMA+ collecting LVs are decreased significantly in the inflamed periodontal tissues. Both mouse models of periodontitis have delayed lymph flow in periodontal tissues, increased TRAP-positive osteoclasts, and significant alveolar bone loss. Importantly, the local administration of adeno-associated virus for vascular endothelial growth factor C, the major growth factor that promotes lymphangiogenesis, increases the area and number of PDPN+ /αSMA+ collecting LVs, promotes local lymphatic drainage, and reduces alveolar bone loss in both models of periodontitis. Lastly, LYVE1+ /αSMA- lymphatic capillaries are increased, whereas LYVE1+ /αSMA+ collecting LVs are decreased significantly in gingival tissues of patients with chronic periodontitis compared with those of clinically healthy controls. Thus, our findings reveal an important role of local lymphatic drainage in periodontal inflammation-mediated alveolar bone loss. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Perda do Osso Alveolar/prevenção & controle , Processo Alveolar/metabolismo , Periodontite Crônica/terapia , Terapia Genética , Linfa/metabolismo , Vasos Linfáticos/metabolismo , Maxila/metabolismo , Fator C de Crescimento do Endotélio Vascular/biossíntese , Fator C de Crescimento do Endotélio Vascular/genética , Perda do Osso Alveolar/genética , Perda do Osso Alveolar/metabolismo , Perda do Osso Alveolar/patologia , Processo Alveolar/patologia , Animais , Estudos de Casos e Controles , Periodontite Crônica/genética , Periodontite Crônica/metabolismo , Periodontite Crônica/patologia , Modelos Animais de Doenças , Humanos , Vasos Linfáticos/patologia , Masculino , Maxila/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoclastos/metabolismo , Osteoclastos/patologia , Fator de Necrose Tumoral alfa/genética
6.
Reprod Sci ; 27(7): 1436-1442, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32016798

RESUMO

Androgen is known to regulate microRNA-135a (miR-135a) and can be regulated by androgen, suggesting that it may contribute to polycystic ovary syndrome (PCOS) with hyperandrogenism. However, its roles and mechanisms of action in PCOS are unknown. In this study, the role and molecular mechanisms underlying miR-135a in granulosa cells (GCs) in PCOS were evaluated. miR-135a expression was upregulated in patients with PCOS and in GCs isolated from patients compared with that in the respective controls (P < 0.01), as determined by RT-qPCR. The overexpression of miR-135a inhibited GC proliferation and induced GC apoptosis, as observed by CCK-8 assay and apoptosis assay. Furthermore, miR-135a overexpression increased the expression of double-strand break maker, γH2AX, as confirmed by western blotting. Our results further suggest that these effects were mediated via downregulation of vascular endothelial growth factor C (VEGFC), which was identified as a direct target of miR-135a. Moreover, levels of VEGFC and miR-135a expression showed a negative correlation. These findings indicate that miR-135a promotes apoptosis and the DNA damage response in GCs in PCOS, likely via VEGFC signaling. This study provides novel insights into GC dysregulation in PCOS and suggests that miR-135a is a promising therapeutic target for PCOS treatment.


Assuntos
Apoptose/fisiologia , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , MicroRNAs/biossíntese , Síndrome do Ovário Policístico/metabolismo , Fator C de Crescimento do Endotélio Vascular/biossíntese , Adulto , Células Cultivadas , Feminino , Expressão Gênica , Células da Granulosa/patologia , Humanos , Células Lúteas/patologia , MicroRNAs/genética , Síndrome do Ovário Policístico/patologia , Fator C de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator C de Crescimento do Endotélio Vascular/genética , Adulto Jovem
7.
Mol Cell Biochem ; 457(1-2): 93-103, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30993496

RESUMO

Metastasis accounts for the majority of cancer-associated mortality and renders the targeted therapy fruitless in the patients of breast cancer. Matrix metalloproteinase-9 (MMP-9) and vascular endothelial growth factor (VEGF-C) are thought to be involved in tumor progression and metastasis. The aim of this study was to investigate the expression of MMP-9 and VEGF-C at both mRNA and protein levels in breast cancer and to correlate with lymph node metastasis and other clinicopathological characteristics. Biopsy specimens (N = 100) of breast cancer & benign breast disease (N = 100) were investigated for the mRNA expression of MMP-9 and VEGF-C by Real-time PCR and Protein expression by Western blot. Elevated levels of MMP-9 (p < 0.001) and VEGF-C (p < 0.001) expression were detected in breast cancer with corresponding to benign breast disease. Additionally, we found significantly increased levels of MMP-9 and VEGF-C in node-positive group with respect to node-negative group. Moreover, the levels of MMP-9 were significantly increased in larger tumor size (T3/T4) (p < 0.05) as compared to smaller size (T1/T2), which suggests that MMP-9 plays an important role in the progression of breast cancer. VEGF-C expression was associated with the TNM stage of tumor (p < 0.05). Further, a significant positive correlation was established between the mRNA levels of these two genes (p < 0.001). However, we could not obtain any significant correlation between expression of these genes with other clinicopathological parameters like tumor grade, age, menopausal status, and receptor status like ER, PR, and Her2. This study suggests that the high expression of MMP-9 and VEGF-C could act as markers for the tumor presence in breast cancer. In addition, this study recommends that expression of MMP-9 and VEGF-C was significantly associated with lymph node status and may provide valuable diagnosis of lymph node metastasis in breast cancer patients. Further, MMP-9 expression was associated with the tumor size and VEGF-C expression was correlated with the staging of the tumor, although no association was observed with other clinicopathological variables.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama , Regulação Neoplásica da Expressão Gênica , Metaloproteinase 9 da Matriz , Proteínas de Neoplasias , Fator C de Crescimento do Endotélio Vascular , Adulto , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Índia , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/genética , Pessoa de Meia-Idade , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Fator C de Crescimento do Endotélio Vascular/biossíntese , Fator C de Crescimento do Endotélio Vascular/genética
8.
EMBO Rep ; 20(4)2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30783017

RESUMO

A lacteal is a blunt-ended, long, tube-like lymphatic vessel located in the center of each intestinal villus that provides a unique route for drainage of absorbed lipids from the small intestine. However, key regulators for maintaining lacteal integrity are poorly understood. Here, we explore whether and how the gut microbiota regulates lacteal integrity. Germ depletion by antibiotic treatment triggers lacteal regression during adulthood and delays lacteal maturation during the postnatal period. In accordance with compromised lipid absorption, the button-like junction between lymphatic endothelial cells, which is ultrastructurally open to permit free entry of dietary lipids into lacteals, is significantly reduced in lacteals of germ-depleted mice. Lacteal defects are also found in germ-free mice, but conventionalization of germ-free mice leads to normalization of lacteals. Mechanistically, VEGF-C secreted from villus macrophages upon MyD88-dependent recognition of microbes and their products is a main factor in lacteal integrity. Collectively, we conclude that the gut microbiota is a crucial regulator for lacteal integrity by endowing its unique microenvironment and regulating villus macrophages in small intestine.


Assuntos
Microbioma Gastrointestinal , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Macrófagos/metabolismo , Fator C de Crescimento do Endotélio Vascular/biossíntese , Fatores Etários , Animais , Transporte Biológico , Biomarcadores , Receptor 1 de Quimiocina CX3C/metabolismo , Imunofluorescência , Absorção Intestinal , Mucosa Intestinal/citologia , Mucosa Intestinal/ultraestrutura , Metabolismo dos Lipídeos , Camundongos , Microvasos/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais
9.
Cell Physiol Biochem ; 52(1): 1-15, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30790501

RESUMO

BACKGROUND/AIMS: Chondrosarcoma is the second most common primary malignancy of bone, characterized by a high metastatic potential. Increasing clinical data highlight the important role played by lymphangiogenesis in cancer metastasis. Amphiregulin (AR) has been implicated in tumor metastasis and lymphangiogenesis, but its association with vascular endothelial growth factor-C (VEGF-C) expression and lymphangiogenesis in chondrosarcoma is unclear. METHODS: We used qPCR, ELISA and Western blotting to detect AR-induced VEGF-C expression in chondrosarcoma cells. Lymphangiogenesis was investigated by lymphatic endothelial cells (LECs) migration and tube formation. An in vivo experiment examined AR expression in tumor-associated lymphangiogenesis. RESULTS: In this study, we found that both AR and VEGF-C expression correlated with tumor stage and were significantly higher than levels found in normal cartilage. Exogenous AR promoted VEGF-C expression in chondrosarcoma cells in a time- and dose-dependent manner and subsequently increased migration and tube formation of LECs. AR also increased VEGF-C expression and lymphangiogenesis through the Src/MEK/ERK/STAT3 signaling pathway. However, it is unclear as to how an EGFR ligand (AR) induces activation of the Src kinase. Knockdown of AR decreased VEGF-C expression in chondrosarcoma cells. Similarly, lymphangiogenesis was abolished in AR knockdown cells in an in vivo model of chondrosarcoma. CONCLUSION: These results indicate that AR occurs through the Src/MEK/ERK/STAT-3 pathway, activating VEGF-C expression and contributing to lymphangiogenesis in human chondrosarcoma. Thus, AR could be a therapeutic target in metastasis and lymphangiogenesis of chondrosarcoma.


Assuntos
Anfirregulina/metabolismo , Neoplasias Ósseas/metabolismo , Condrossarcoma/metabolismo , Regulação Neoplásica da Expressão Gênica , Linfangiogênese , Proteínas de Neoplasias/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator C de Crescimento do Endotélio Vascular/biossíntese , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Condrossarcoma/patologia , Humanos
10.
J Hepatol ; 70(4): 700-709, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30553841

RESUMO

BACKGROUND & AIMS: In cholangiocarcinoma, early metastatic spread via lymphatic vessels often precludes curative therapies. Cholangiocarcinoma invasiveness is fostered by an extensive stromal reaction, enriched in cancer-associated fibroblasts (CAFs) and lymphatic endothelial cells (LECs). Cholangiocarcinoma cells recruit and activate CAFs by secreting PDGF-D. Herein, we investigated the role of PDGF-D and liver myofibroblasts in promoting lymphangiogenesis in cholangiocarcinoma. METHODS: Human cholangiocarcinoma specimens were immunostained for podoplanin (LEC marker), α-SMA (CAF marker), VEGF-A, VEGF-C, and their cognate receptors (VEGFR2, VEGFR3). VEGF-A and VEGF-C secretion was evaluated in human fibroblasts obtained from primary sclerosing cholangitis explants. Using human LECs incubated with conditioned medium from PDGF-D-stimulated fibroblasts we assessed migration, 3D vascular assembly, transendothelial electric resistance and transendothelial migration of cholangiocarcinoma cells (EGI-1). We then studied the effects of selective CAF depletion induced by the BH3 mimetic navitoclax on LEC density and lymph node metastases in vivo. RESULTS: In cholangiocarcinoma specimens, CAFs and LECs were closely adjacent. CAFs expressed VEGF-A and VEGF-C, while LECs expressed VEGFR2 and VEGFR3. Upon PDGF-D stimulation, fibroblasts secreted increased levels of VEGF-C and VEGF-A. Fibroblasts, stimulated by PDGF-D induced LEC recruitment and 3D assembly, increased LEC monolayer permeability, and promoted transendothelial EGI-1 migration. These effects were all suppressed by the PDGFRß inhibitor, imatinib. In the rat model of cholangiocarcinoma, navitoclax-induced CAF depletion, markedly reduced lymphatic vascularization and reduced lymph node metastases. CONCLUSION: PDGF-D stimulates VEGF-C and VEGF-A production by fibroblasts, resulting in expansion of the lymphatic vasculature and tumor cell intravasation. This critical process in the early metastasis of cholangiocarcinoma may be blocked by inducing CAF apoptosis or by inhibiting the PDGF-D-induced axis. LAY SUMMARY: Cholangiocarcinoma is a highly malignant cancer affecting the biliary tree, which is characterized by a rich stromal reaction involving a dense population of cancer-associated fibroblasts that promote early metastatic spread. Herein, we show that cholangiocarcinoma-derived PDGF-D stimulates fibroblasts to secrete vascular growth factors. Thus, targeting fibroblasts or PDGF-D-induced signals may represent an effective tool to block tumor-associated lymphangiogenesis and reduce the invasiveness of cholangiocarcinoma.


Assuntos
Neoplasias dos Ductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Fígado/patologia , Linfangiogênese/efeitos dos fármacos , Linfocinas/metabolismo , Linfocinas/farmacologia , Miofibroblastos/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Animais , Neoplasias dos Ductos Biliares/patologia , Fibroblastos Associados a Câncer/metabolismo , Linhagem Celular Tumoral , Colangiocarcinoma/patologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Xenoenxertos , Humanos , Mesilato de Imatinib/farmacologia , Masculino , Camundongos , Camundongos SCID , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Endogâmicos F344 , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator C de Crescimento do Endotélio Vascular/biossíntese
11.
Curr Mol Med ; 18(5): 273-286, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30289073

RESUMO

BACKGROUND: Whereas retinal pigment epithelial (RPE) cells are known to secrete VEGF-A and VEGFR2, the functions of the autocrine VEGF signaling remain unclear. Meanwhile, anti-VEGF therapies have been applied routinely to treat ocular vascular diseases. OBJECTIVE: The aim of this study was to determine the functions of the VEGF signaling in RPE cells and evaluate the consequences of its interruption. METHODS: The genes involved in the VEGF and Hippo signal pathways were knocked down with siRNAs in both ARPE-19 cell line and human primary RPE cells via transient transfection whereas overexpression of VEGFR2 was mediated via adenovirus transduction. Expression of the epithelial-mesenchymal transition (EMT) markers and the downstream genes of YAP were determined by real-time PCR and Western Blot analysis. Immunofluorescence staining was utilized to determine gene expression in tissue and mouse samples. RESULTS: Knockdown of VEGFR2 results in epithelial-mesenchymal transition in vitro and in vivo. Overexpression of VEGFR2 suppresses TGF ß-mediated EMT in RPE cells. Loss of VEGF-C rather than VEGF-A induces EMT. Mechanistically, the VEGFR2 ablation-induced EMT in RPE cells is mediated by activation of YAP, an effector of the Hippo pathway. Finally, the immunohistochemical analysis of VEGFR2 and YAP in human proliferative vitreoretinopathy (PVR) membranes indicates a tendency of an inverse correlation between VEGFR2-positive and YAP-positive cells. CONCLUSIONS: Our results disclose unexpected novel roles of VEGFR2 and VEGF-C in the process of EMT of RPE cells and in the Hippo pathway. The data shown here demonstrated that VEGFR2 and VEGF-C are important to maintain the normal physiological state of RPE cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica , Fosfoproteínas/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Transdução de Sinais , Fator C de Crescimento do Endotélio Vascular/biossíntese , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular , Técnicas de Silenciamento de Genes , Humanos , Fosfoproteínas/genética , Epitélio Pigmentado da Retina/citologia , Fatores de Transcrição , Fator C de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Proteínas de Sinalização YAP
12.
Development ; 145(10)2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29773646

RESUMO

Despite the essential role of the lymphatic vasculature in tissue homeostasis and disease, knowledge of the organ-specific origins of lymphatic endothelial progenitor cells remains limited. The assumption that most murine embryonic lymphatic endothelial cells (LECs) are venous derived has recently been challenged. Here, we show that the embryonic dermal blood capillary plexus constitutes an additional, local source of LECs that contributes to the formation of the dermal lymphatic vascular network. We describe a novel mechanism whereby rare PROX1-positive endothelial cells exit the capillary plexus in a Ccbe1-dependent manner to establish discrete LEC clusters. As development proceeds, these clusters expand and further contribute to the growing lymphatic system. Lineage tracing and analyses of Gata2-deficient mice confirmed that these clusters are endothelial in origin. Furthermore, ectopic expression of Vegfc in the vasculature increased the number of PROX1-positive progenitors within the capillary bed. Our work reveals a novel source of lymphatic endothelial progenitors employed during construction of the dermal lymphatic vasculature and demonstrates that the blood vasculature is likely to remain an ongoing source of LECs during organogenesis, raising the question of whether a similar mechanism operates during pathological lymphangiogenesis.


Assuntos
Capilares/citologia , Células Endoteliais/citologia , Proteínas de Homeodomínio/genética , Linfangiogênese/fisiologia , Vasos Linfáticos/embriologia , Células-Tronco/citologia , Proteínas Supressoras de Tumor/genética , Animais , Proteínas de Ligação ao Cálcio/genética , Fator de Transcrição GATA2/genética , Linfangiogênese/genética , Vasos Linfáticos/citologia , Camundongos , Camundongos Transgênicos , Fator C de Crescimento do Endotélio Vascular/biossíntese , Fator C de Crescimento do Endotélio Vascular/genética
13.
Biochem Pharmacol ; 154: 234-242, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29730230

RESUMO

Chondrosarcoma is a common primary malignant tumor of the bone that can metastasize through the vascular system to other organs. A key step in the metastatic process, lymphangiogenesis, involves vascular endothelial growth factor-C (VEGF-C). However, the effects of lymphangiogenesis in chondrosarcoma metastasis remain to be clarified. Accumulating evidence shows that resistin, a cytokine secreted from adipocytes and monocytes, also promotes tumor pathogenesis. Notably, chondrosarcoma can easily metastasize. In this study, we demonstrate that resistin enhances VEGF-C expression and lymphatic endothelial cells (LECs)-associated lymphangiogenesis in human chondrosarcoma cells. We also show that resistin triggers VEGF-C-dependent lymphangiogenesis via the c-Src signaling pathway and down-regulating micro RNA (miR)-186. Overexpression of resistin in chondrosarcoma cells significantly enhanced VEGF-C production and LECs-associated lymphangiogenesis in vitro and tumor-related lymphangiogenesis in vivo. Resistin levels were positively correlated with VEGF-C-dependent lymphangiogenesis via the down-regulation of miR-186 expression in clinical samples from chondrosarcoma tissue. This study is the first to evaluate the mechanism underlying resistin-induced promotion of LECs-associated lymphangiogenesis via the upregulation of VEGF-C expression in human chondrosarcomas. We suggest that resistin may represent a molecular target in VEGF-C-associated tumor lymphangiogenesis in chondrosarcoma metastasis.


Assuntos
Neoplasias Ósseas/metabolismo , Condrossarcoma/metabolismo , Linfangiogênese/fisiologia , MicroRNAs/biossíntese , Resistina/biossíntese , Fator C de Crescimento do Endotélio Vascular/biossíntese , Neoplasias Ósseas/genética , Linhagem Celular Transformada , Linhagem Celular Tumoral , Condrossarcoma/genética , Relação Dose-Resposta a Droga , Humanos , Linfangiogênese/efeitos dos fármacos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Resistina/genética , Resistina/farmacologia , Fator C de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator C de Crescimento do Endotélio Vascular/genética
14.
Prostate ; 78(9): 682-690, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29601651

RESUMO

BACKGROUND: Metastasis is the major cause of cancer-specific death in patients with prostate cancer (PCa). We previously reported that collapsing response mediator protein-4 (CRMP4) is a PCa metastasis-suppressor gene and the hypermethylation in CRMP4 promoter is responsible for the transcription repression in metastatic PCa. However, the underlying mechanisms remain unknown. In this study, we aimed to investigate the role of calpain-2 in CRMP4 promoter hypermethylation and its functional modulation in PCa metastasis. METHODS: Calpain-2 expression in PCa tissues (n = 87) and its specific mechanisms of functional modulation in CRMP4 expression via limited enzymatic cleavage was investigated. We then focused on the cooperative crosstalk of calpain-2 and NF-κB RelA/p65 in CRMP4 promoter methylation for the initiation of PCa metastasis. Statistical differences between groups were determined using a two-tailed Student's t-test. P < 0.05 indicated statistically significant. RESULTS: Calpain-2 was differentially upregulated in metastatic PCa compared with localized PCa. Moreover, calpain-2 cleaved CRMP4 into the N-terminally fragment which promoted migration and invasion in PCa cells via nuclear translocation and activation of E2F1-mediated DNA methyltransferase 1 (DNMT1) expression. NF-κB RelA/p65 recruited DNMT1 to bind to and methylate CRMP4 promoter in which Serine276 phosphorylation of p65 was essential. Furthermore, CRMP4 exhibited anti-metastatic function via inhibiting the expression of VEGFC through Semaphorin3B-Neuropilin2 signaling. CONCLUSION: Calpain-2 may contribute to the promoter methylation of CRMP4 to repress its transcription, leading to the metastasis of PCa via enhancing VEGFC expression.


Assuntos
Calpaína/biossíntese , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Proteínas Musculares/metabolismo , Metástase Neoplásica/fisiopatologia , Neoplasias da Próstata/metabolismo , Fator de Transcrição RelA/metabolismo , Idoso , Linhagem Celular Tumoral , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferase 1/biossíntese , Metilação de DNA , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Supressores de Tumor/fisiologia , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Proteínas Musculares/biossíntese , Metástase Neoplásica/genética , Neuropilina-2/metabolismo , Fosforilação , Regiões Promotoras Genéticas/fisiologia , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/secundário , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/secundário , Receptor Cross-Talk/fisiologia , Estudos Retrospectivos , Semaforinas/metabolismo , Transdução de Sinais , Regulação para Cima , Fator C de Crescimento do Endotélio Vascular/biossíntese
15.
Heart Vessels ; 33(9): 1106-1120, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29557990

RESUMO

The vascular endothelial growth factor (VEGF) family of peptides and caveolins (CAVs) are reported to contribute, in early graft failure in patients, a coronary artery bypass grafting (CABG). To investigate the possible association of ultimate luminal occlusion to VEGFs and CAVs expression, a functional analysis (based on the molecular biology, bioinformatics, histology, and clinical studies) was performed. Twenty-four hundred and sixty-eight CABG patients diagnosed with multivessel stable coronary artery disease (CAD) were enrolled into prospective study and assigned to two subgroups: double- and triple-vessel CAD subjects. Distal parts of all the harvested saphenous vein (SV) and internal thoracic artery (ITA) segments were used for further tests. ITA graft failure did not differ between double-vessel and triple-vessel CAD patients. The number of SV occlusions was significantly higher in triple-vessel CAD subjects. The microarray analysis performed on SV and ITA samples obtained exclusively from triple-vessel CAD patients who developed early graft occlusion revealed 383 genes with increased and 301 genes with decreased expression in ITA samples as compared to SV grafts. This was followed by functional analysis of 'blood vessel development' group of genes. Average VEGF-C expression in ITA grafts was higher than in corresponding SV grafts; FLT4 expression was significantly higher in SV than in ITA transplants. VEGFR-3 and CAV3 expression demonstrated immunohistochemically in SMCs of the tunica media of SV grafts predicted their early restenosis in triple-vessel CAD patients. CAV2 protein expression in SMCs of ITA grafts indicated the risk of early graft failure both in double-vessel and triple-vessel CAD subjects.


Assuntos
Ponte de Artéria Coronária/efeitos adversos , Doença da Artéria Coronariana/cirurgia , Vasos Coronários/cirurgia , Regulação da Expressão Gênica , Oclusão de Enxerto Vascular/genética , Fator C de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Idoso , Angiografia Coronária , Vasos Coronários/diagnóstico por imagem , Vasos Coronários/fisiopatologia , Feminino , Oclusão de Enxerto Vascular/diagnóstico , Oclusão de Enxerto Vascular/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Tomografia Computadorizada Multidetectores , Estudos Prospectivos , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Resultado do Tratamento , Fator C de Crescimento do Endotélio Vascular/biossíntese , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Grau de Desobstrução Vascular
16.
Int J Oncol ; 51(6): 1920-1928, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29075785

RESUMO

Lymph node metastasis is one of the most valuable determinants for the prognosis of ovarian cancer. However, the molecular mechanisms underlying lymphangiogenesis in ovarian cancer is still poorly understood. Secreted protein acidic and rich in cysteine (SPARC), a Ca2+-binding matricellular glycoprotein that modulates cell adhesion, migration and differentiation, is thought to play a decisive role in tumor metastasis. Vascular endothelial growth factor (VEGF)-C and VEGF-D contributes to tumor-associated lymphatic vessel growth, enhancing the metastatic spread of tumor cells to lymph nodes. The aim of the present study was to investigate the relationship among SPARC, VEGFs and lymph node metastasis in ovarian cancer. We found that SKOV3 cells expressed high-level SPARC, much more than SKOV3-PM4 cells (a subline with high directional lymphatic metastatic potentials established from the metastatic lymph node generated by human ovarian carcinoma cell line SKOV3 in nude mice) did at both mRNA and protein levels. A SPARC-overexpressed SKOV3-PM4 cell line was constructed and it was found that upregulation of SPARC expression suppressed the growth, migration and invasion of SKOV3-PM4 cells as well as markedly reduced the expression of VEGF-D at both mRNA and protein level by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot assay. In 47 of ovarian malignant tissues, the expression of SPARC, VEGF-C and VEGF-D were determined by immunohistochemistry. Lymphatic microvessel density (LVD) and microvessel density (MVD) were evaluated by immunostaining with CD34 and D2-40 antibodies, respectively. We found that SPARC expression was significantly lower in tissues with lymph node metastasis as compared to tissues without lymph node metastasis. SPARC expression was inversely associated with the degree of malignancy and it had a negative correlation with VEGF-C expression, VEGF-D expression, LVD and MVD which were actually higher for advanced tumors than for non-advanced tumors. These results suggest SPARC might function as a tumor suppressor inhibiting angiogenesis and lymphangiogenesis in ovarian cancer by reducing the expression of VEGF-C and VEGF-D.


Assuntos
Osteonectina/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fator C de Crescimento do Endotélio Vascular/biossíntese , Fator D de Crescimento do Endotélio Vascular/biossíntese , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Feminino , Humanos , Metástase Linfática , Microscopia Confocal , Neovascularização Patológica/patologia , Osteonectina/genética , Neoplasias Ovarianas/irrigação sanguínea , Transfecção , Fator C de Crescimento do Endotélio Vascular/genética , Fator D de Crescimento do Endotélio Vascular/genética
17.
Melanoma Res ; 27(6): 565-572, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28984690

RESUMO

Regional lymph nodes are affected frequently by melanoma metastasis. Its microenvironment may be associated with tumor progression. We investigated sentinel nodes with and without tumor and negative nodes surrounding positive nodes, looking for patterns related to tumor immune interaction and lymphovascular progression. We quantified programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1, vascular endothelial growth factor (VEGF)-A/VEGF-C expressions in lymph nodes of 103 patients who underwent sentinel lymph node biopsy. Two groups were studied: negative sentinel lymph nodes and positive ones. Negative lymph nodes of sequential lymphadenectomy from positive cases were also studied. Markers were assessed by immunohistochemistry. Results were related to clinical/histological outcomes. VEGF-A/VEGF-C analysis showed higher positivity in metastatic nodes and higher positivity in the surrounding negative nodes from positive cases in comparison with nonmetastatic patients. Programmed cell death-ligand 1, studied only in metastasis, presented high positivity, not associated with prognosis. PD-1 expressions were similar in the groups with a 1% cutoff and higher in the metastasis with a 5% cutoff. Higher VEGF-A expression was related to higher pathological stages. PD-1 expression in the lymph node was associated with higher survival. Other clinical and histopatological variables were not associated with marker expression patterns. VEGF-A and VEGF-C expressions in lymph nodes were associated with the presence of lymph node metastasis. PD-1 expression in the lymph node was related to higher survival rates and this should be explored in the context of adjuvant immunotherapy.


Assuntos
Antígeno B7-H1/biossíntese , Linfonodos/metabolismo , Melanoma/metabolismo , Receptor de Morte Celular Programada 1/biossíntese , Neoplasias Cutâneas/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator C de Crescimento do Endotélio Vascular/biossíntese , Adulto , Feminino , Humanos , Imuno-Histoquímica , Linfonodos/patologia , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Metástase Neoplásica , Estudos Retrospectivos , Neoplasias Cutâneas/patologia , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
18.
Circulation ; 136(17): 1629-1642, 2017 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-28860123

RESUMO

BACKGROUND: The vascular complications of diabetes mellitus are the major causes of morbidity and mortality among people with diabetes. Circular RNAs are a class of endogenous noncoding RNAs that regulate gene expression in eukaryotes. In this study, we investigated the role of circular RNA in retinal vascular dysfunction induced by diabetes mellitus. METHODS: Quantitative polymerase chain reactions, Sanger sequencing, and Northern blots were conducted to detect circular HIPK3 (circHIPK3) expression pattern on diabetes mellitus-related stresses. MTT (3-[4,5-dimethythiazol-2-yl]-2,5-diphenyl tetrazolium bromide) assays, EdU (5-ethynyl-2'-deoxyuridine) incorporation assays, Transwell migration assays, and Matrigel assays were conducted to detect the role of circHIPK3 in retinal endothelial cell function in vitro. Retinal trypsin digestion, vascular permeability assays, and ELISA assays were conducted to detect the role of circHIPK3 in retinal vascular dysfunction in vivo. Bioinformatics analysis, luciferase activity assays, RNA pull-down assays, and in vitro studies were conducted to reveal the mechanism of circHIPK3-mediated retinal vascular dysfunction. RESULTS: circHIPK3 expression was significantly upregulated in diabetic retinas and retinal endothelial cells following stressors related to diabetes mellitus. circHIPK3 silencing or overexpressing circHIPK3 changed retinal endothelial cell viability, proliferation, migration, and tube formation in vitro. circHIPK3 silencing in vivo alleviated retinal vascular dysfunction, as shown by decreased retinal acellular capillaries, vascular leakage, and inflammation. circHIPK3 acted as an endogenous miR-30a-3p sponge to sequester and inhibit miR-30a-3p activity, which led to increased vascular endothelial growth factor-C, FZD4, and WNT2 expression. Ectopic expression of miR-30a-3p mimicked the effect of circHIPK3 silencing on vascular endothelial phenotypes in vivo and in vitro. CONCLUSIONS: The circular RNA circHIPK3 plays a role in diabetic retinopathy by blocking miR-30a function, leading to increased endothelial proliferation and vascular dysfunction. These data suggest that circular RNA is a potential target to control diabetic proliferative retinopathy.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/metabolismo , Células Endoteliais/metabolismo , RNA não Traduzido/metabolismo , Vasos Retinianos/metabolismo , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Células Endoteliais/fisiologia , Receptores Frizzled/biossíntese , Receptores Frizzled/genética , Regulação da Expressão Gênica , Masculino , Camundongos , MicroRNAs/biossíntese , MicroRNAs/genética , RNA não Traduzido/genética , Vasos Retinianos/patologia , Fator C de Crescimento do Endotélio Vascular/biossíntese , Fator C de Crescimento do Endotélio Vascular/genética , Proteínas Wnt/biossíntese , Proteínas Wnt/genética
19.
Cell Death Dis ; 8(8): e2964, 2017 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-28771226

RESUMO

Chondrosarcoma is the second most common primary malignancy of bone, and one of the most difficult bone tumors to diagnose and treat. It is well known that increased levels of vascular endothelial growth factor-C (VEGF-C) promote active tumor lymphangiogenesis and lymphatic tumor spread to regional lymph nodes. Brain-derived neurotrophic factor (BDNF) is known to promote metastasis in human chondrosarcoma cells. Knowing more about the mechanism of BDNF in VEGF-C expression and lymphangiogenesis in human chondrosarcoma would improve our understanding as how to prevent chondrosarcoma angiogenesis and metastasis, which currently lacks effective adjuvant treatment. Here, we found that BDNF expression was at least 2.5-fold higher in the highly migratory JJ012(S10) cell line as compared with the primordial cell line (JJ012). In addition, VEGF-C expression and secretion was markedly increased in JJ012(S10) cells. Conditioned medium from JJ012(S10) cells significantly promoted migration and tube formation of human lymphatic endothelial cells (LECs), whereas knockdown of BDNF attenuated LEC migration and tube formation by suppressing VEGF-C production in JJ012(S10) cells. Mechanistic investigations indicated that BDNF facilitated VEGF-C-dependent lymphangiogenesis through the MEK/ERK/mTOR signaling pathway. We also showed that microRNA (miR)-624-3p expression was negatively regulated by BDNF via the MEK/ERK/mTOR cascade. Importantly, BDNF knockdown profoundly inhibited tumor-associated lymphangiogenesis in vivo. Further analyses identified that BDNF promoted tumor lymphangiogenesis by downregulating miR-624-3p in human chondrosarcoma tissues. In conclusion, this study is the first to reveal the mechanism underlying BDNF-induced lymphangiogenesis. We suggest that BDNF may serve as a promising therapeutic target for the restriction of VEGF-C-mediated tumor lymphangiogenesis and lymphatic metastasis.


Assuntos
Neoplasias Ósseas/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Condrossarcoma/metabolismo , Linfangiogênese/efeitos dos fármacos , MicroRNAs/biossíntese , Proteínas de Neoplasias/biossíntese , RNA Neoplásico/biossíntese , Fator C de Crescimento do Endotélio Vascular/biossíntese , Adulto , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Condrossarcoma/genética , Condrossarcoma/patologia , Feminino , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , RNA Neoplásico/genética
20.
J Plast Reconstr Aesthet Surg ; 70(11): 1653-1659, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28756123

RESUMO

INTRODUCTION: In many patients, the clinical behaviour of cutaneous melanoma is very difficult to predict by traditional histologic and clinical parameters. This study aimed to examine the role of quantitative parameters of tumour lymphangiogenesis and vascular endothelial growth factor (VEGF)-C in predicting metastatic risk in patients with cutaneous melanoma. METHODS: One hundred melanoma specimens were stained with lymphatic-specific antibody D2-40 and with anti-VEGF-C antibody. Quantitative parameters of lymphangiogenesis-lymphatic vessel density (LVD) and lymphatic vessel area (LVA)-were determined by computer-assisted morphometric analysis. Moderate or strong staining was assessed as a positive expression of VEGF-C in tumour cells. RESULTS: Univariate analysis revealed that intratumoural LVD, peritumoural LVD, VEGF-C expression in tumour cells, melanoma thickness, Clark level, ulceration, gender and histologic type were significant predictors of lymph node metastasis (p = 0.000, p = 0.000, p = 0.000, p = 0.000, p = 0.005, p = 0.005, p = 0.011 and p = 0.027, respectively). No significant association of intratumoural and peritumoural LVA with metastases was found. In multivariate analysis, independent predictors of metastatic risks were melanoma thickness [odds ratio OR = 1.655, 95% confidence interval (CI) 1.102-2.484, p = 0.015], intratumoural LVD (OR = 1.086, 95% CI 1.027-1.148, p = 0.004), peritumoural LVD (OR = 1.050, 95% CI 1.008-1.094, p = 0.020) and a positive VEGF-C expression in tumour cells (OR = 20.337, 95% CI 2.579-160.350, p = 0.004). CONCLUSIONS: This study identified intratumoural and peritumoural LVD and the VEGF-C expression in tumour cells as more significant predictors of metastatic risk than melanoma thickness, ulceration and other clinical-pathological parameters.


Assuntos
Vasos Linfáticos/patologia , Melanoma/secundário , Neoplasias Cutâneas/secundário , Fator C de Crescimento do Endotélio Vascular/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/biossíntese , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Linfangiogênese , Metástase Linfática , Masculino , Melanoma/diagnóstico , Melanoma/metabolismo , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...