Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Pathol ; 236(4): 491-504, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25880038

RESUMO

Epithelial-mesenchymal transition (EMT) programmes provide cancer cells with invasive and survival capacities that might favour metastatic dissemination. Whilst signalling cascades triggering EMT have been extensively studied, the impact of EMT on the crosstalk between tumour cells and the tumour microenvironment remains elusive. We aimed to identify EMT-regulated soluble factors that facilitate the recruitment of host cells in the tumour. Our findings indicate that EMT phenotypes relate to the induction of a panel of secreted mediators, namely IL-8, IL-6, sICAM-1, PAI-1 and GM-CSF, and implicate the EMT-transcription factor Snail as a regulator of this process. We further show that EMT-derived soluble factors are pro-angiogenic in vivo (in the mouse ear sponge assay), ex vivo (in the rat aortic ring assay) and in vitro (in a chemotaxis assay). Additionally, conditioned medium from EMT-positive cells stimulates the recruitment of myeloid cells. In a bank of 40 triple-negative breast cancers, tumours presenting features of EMT were significantly more angiogenic and infiltrated by a higher quantity of myeloid cells compared to tumours with little or no EMT. Taken together, our results show that EMT programmes trigger the expression of soluble mediators in cancer cells that stimulate angiogenesis and recruit myeloid cells in vivo, which might in turn favour cancer spread.


Assuntos
Proteínas Angiogênicas/metabolismo , Quimiotaxia , Citocinas/metabolismo , Transição Epitelial-Mesenquimal , Células Mieloides/metabolismo , Neovascularização Patológica , Comunicação Parácrina , Neoplasias de Mama Triplo Negativas/irrigação sanguínea , Neoplasias de Mama Triplo Negativas/metabolismo , Microambiente Tumoral , Proteínas Angiogênicas/genética , Animais , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/metabolismo , Citocinas/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Camundongos SCID , Células Mieloides/patologia , Fenótipo , Interferência de RNA , Ratos , Transdução de Sinais , Transfecção , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
2.
J Pathol ; 234(4): 464-77, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25196670

RESUMO

Human papillomavirus (HPV) infection, particularly type 16, is causally associated with cancer of the uterine cervix, which mainly develops at the squamocolumnar (SC) junction. The progression of cervical HPV infections into (pre)neoplastic lesions suggests that viral antigens are not adequately recognized by innate immunity or presented to the adaptive immune system. Members of the defensin family have recently been found to inhibit viral and bacterial pathogens, to stimulate the migration of immune cells and to play a role in anticancer responses. In the present study, we focused on the poorly characterized human α-defensin 5 (HD-5) and its possible role in these processes. We showed that HD-5 was able to prevent HPV virion entry into cervical keratinocytes and to influence adaptive immunity. Indeed, this peptide specifically induced the chemoattraction and proliferation of both activated T lymphocytes and immature dendritic cells in a CCR2/CCR6-dependent manner and stimulated the infiltration of these professional antigen-presenting cells in a (pre)neoplastic epithelium transplanted in vivo in immunodeficient mice. No chemotactic effect was observed with plasmacytoid dendritic cells, macrophages or natural killer cells. Proliferative and angiogenic effects of HD-5 were also assessed in vitro and in vivo. However there was a striking regional disparity in expression of HD-5, being prominent in ectocervical, vaginal and vulvar neoplasia, while absent, or nearly so, in the cervical SC junction. Taken together, these results suggest one possible explanation for why the SC junction is uniquely vulnerable to both high-risk HPV infection (via reduced HD-5 expression and viral entry) and progression of neoplasia (via altered cell-mediated immune responses and altered microenvironment).


Assuntos
Colo do Útero/metabolismo , Infecções por Papillomavirus/imunologia , Lesões Pré-Cancerosas/imunologia , Neoplasias Uterinas/virologia , alfa-Defensinas/biossíntese , Animais , Western Blotting , Células Cultivadas , Colo do Útero/imunologia , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Xenoenxertos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/virologia , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias Uterinas/imunologia , Displasia do Colo do Útero/imunologia , Displasia do Colo do Útero/virologia
3.
J Biol Chem ; 288(15): 10195-204, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23413031

RESUMO

The group of matrix metalloproteases (MMPs) is responsible for multiple processes of extracellular matrix remodeling in the healthy body but also for matrix and tissue destruction during cancer invasion and metastasis. The understanding of the contributions from each individual MMP, both in healthy and pathological events, has been complicated by the lack of specific inhibitors and the fact that some of the potent MMPs are multifunctional enzymes. These factors have also hampered the setup of therapeutic strategies targeting MMP activity. A tempting target is the membrane-associated MT1-MMP, which has well-documented importance in matrix degradation but which takes part in more than one pathway in this regard. In this report, we describe the selective targeting of a single function of this enzyme by means of a specific monoclonal antibody against MT1-MMP, raised in an MT1-MMP knock-out mouse. The antibody blocks the enzyme ability to activate proMMP-2 without interfering with the collagenolytic function or the general proteolytic activity of MT1-MMP. Using this antibody, we have shown that the MT1-MMP-catalyzed activation of proMMP-2 is involved in the outgrowth of cultured lymphatic endothelial cells in a collagen matrix in vitro, as well as in lymphatic vessel sprouting assayed ex vivo. This is the first example of the complete inactivation of a single function of a multifunctional MMP and the use of this strategy to pursue its role.


Assuntos
Matriz Extracelular/metabolismo , Linfangiogênese/fisiologia , Metaloproteinase 14 da Matriz/metabolismo , Animais , Anticorpos Monoclonais Murinos/química , Células CHO , Cricetinae , Ativação Enzimática/genética , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/patologia , Gelatinases/genética , Gelatinases/metabolismo , Humanos , Metaloproteinase 14 da Matriz/genética , Camundongos , Camundongos Knockout , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo
4.
Mol Cancer ; 13: 108, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24886454

RESUMO

BACKGROUND: DUSP3 phosphatase, also known as Vaccinia-H1 Related (VHR) phosphatase, encoded by DUSP3/Dusp3 gene, is a relatively small member of the dual-specificity protein phosphatases. In vitro studies showed that DUSP3 is a negative regulator of ERK and JNK pathways in several cell lines. On the other hand, DUSP3 is implicated in human cancer. It has been alternatively described as having tumor suppressive and oncogenic properties. Thus, the available data suggest that DUSP3 plays complex and contradictory roles in tumorigenesis that could be cell type-dependent. Since most of these studies were performed using recombinant proteins or in cell-transfection based assays, the physiological function of DUSP3 has remained elusive. RESULTS: Using immunohistochemistry on human cervical sections, we observed a strong expression of DUSP3 in endothelial cells (EC) suggesting a contribution for this phosphatase to EC functions. DUSP3 downregulation, using RNA interference, in human EC reduced significantly in vitro tube formation on Matrigel and spheroid angiogenic sprouting. However, this defect was not associated with an altered phosphorylation of the documented in vitro DUSP3 substrates, ERK1/2, JNK1/2 and EGFR but was associated with an increased PKC phosphorylation. To investigate the physiological function of DUSP3, we generated Dusp3-deficient mice by homologous recombination. The obtained DUSP3-/- mice were healthy, fertile, with no spontaneous phenotype and no vascular defect. However, DUSP3 deficiency prevented neo-vascularization of transplanted b-FGF containing Matrigel and LLC xenograft tumors as evidenced by hemoglobin (Hb) and FITC-dextran quantifications. Furthermore, we found that DUSP3 is required for b-FGF-induced microvessel outgrowth in the aortic ring assay. CONCLUSIONS: All together, our data identify DUSP3 as a new important player in angiogenesis.


Assuntos
Carcinoma Pulmonar de Lewis/genética , Fosfatase 3 de Especificidade Dupla/genética , Neovascularização Fisiológica/genética , Animais , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Movimento Celular , Colo do Útero/irrigação sanguínea , Colo do Útero/metabolismo , Colo do Útero/patologia , Colágeno , Combinação de Medicamentos , Fosfatase 3 de Especificidade Dupla/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fatores de Crescimento de Fibroblastos , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Laminina , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Knockout , Neovascularização Patológica/prevenção & controle , Fosforilação , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteoglicanas , Transdução de Sinais
5.
Blood ; 119(21): 5048-56, 2012 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-22490679

RESUMO

Lymphatic dysfunctions are associated with several human diseases, including lymphedema and metastatic spread of cancer. Although it is well recognized that lymphatic capillaries attach directly to interstitial matrix mainly composed of fibrillar type I collagen, the interactions occurring between lymphatics and their surrounding matrix have been overlooked. In this study, we demonstrate how matrix metalloproteinase (MMP)-2 drives lymphatic morphogenesis through Mmp2-gene ablation in mice, mmp2 knockdown in zebrafish and in 3D-culture systems, and through MMP2 inhibition. In all models used in vivo (3 murine models and thoracic duct development in zebrafish) and in vitro (lymphatic ring and spheroid assays), MMP2 blockage or down-regulation leads to reduced lymphangiogenesis or altered vessel branching. Our data show that lymphatic endothelial cell (LEC) migration through collagen fibers is affected by physical matrix constraints (matrix composition, density, and cross-linking). Transmission electron microscopy and confocal reflection microscopy using DQ-collagen highlight the contribution of MMP2 to mesenchymal-like migration of LECs associated with collagen fiber remodeling. Our findings provide new mechanistic insight into how LECs negotiate an interstitial type I collagen barrier and reveal an unexpected MMP2-driven collagenolytic pathway for lymphatic vessel formation and morphogenesis.


Assuntos
Linfangiogênese/genética , Vasos Linfáticos/embriologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/fisiologia , Animais , Animais Geneticamente Modificados , Células Cultivadas , Colágeno Tipo I/metabolismo , Colagenases/genética , Colagenases/metabolismo , Colagenases/fisiologia , Embrião não Mamífero , Líquido Extracelular/enzimologia , Líquido Extracelular/metabolismo , Feminino , Humanos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/fisiologia , Masculino , Metaloproteinase 2 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peixe-Zebra
6.
BMC Cell Biol ; 12: 29, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21702933

RESUMO

BACKGROUND: Abnormal lymphatic vessel formation (lymphangiogenesis) is associated with different pathologies such as cancer, lymphedema, psoriasis and graft rejection. Lymphatic vasculature displays distinctive features than blood vasculature, and mechanisms underlying the formation of new lymphatic vessels during physiological and pathological processes are still poorly documented. Most studies on lymphatic vessel formation are focused on organism development rather than lymphangiogenic events occurring in adults. We have here studied lymphatic vessel formation in two in vivo models of pathological lymphangiogenesis (corneal assay and lymphangioma). These data have been confronted to those generated in the recently set up in vitro model of lymphatic ring assay. Ultrastructural analyses through Transmission Electron Microscopy (TEM) were performed to investigate tube morphogenesis, an important differentiating process observed during endothelial cell organization into capillary structures. RESULTS: In both in vivo models (lymphangiogenic corneal assay and lymphangioma), migrating lymphatic endothelial cells extended long processes exploring the neighboring environment and organized into cord-like structures. Signs of intense extracellular matrix remodeling were observed extracellularly and inside cytoplasmic vacuoles. The formation of intercellular spaces between endothelial cells led to tube formation. Proliferating lymphatic endothelial cells were detected both at the tips of sprouting capillaries and inside extending sprouts. The different steps of lymphangiogenesis observed in vivo are fully recapitulated in vitro, in the lymphatic ring assay and include: (1) endothelial cell alignment in cord like structure, (2) intracellular vacuole formation and (3) matrix degradation. CONCLUSIONS: In this study, we are providing evidence for lymphatic vessel formation through tunneling relying on extensive matrix remodeling, migration and alignment of sprouting endothelial cells into tubular structures. In addition, our data emphasize the suitability of the lymphatic ring assay to unravel mechanisms underlying lymphangiogenesis.


Assuntos
Linfangiogênese/fisiologia , Vasos Linfáticos/fisiologia , Animais , Movimento Celular , Células Cultivadas , Córnea/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Linfático/metabolismo , Endotélio Linfático/patologia , Feminino , Adjuvante de Freund , Hiperplasia , Técnicas In Vitro , Linfangioma/etiologia , Linfangioma/patologia , Vasos Linfáticos/patologia , Vasos Linfáticos/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão
7.
Nat Commun ; 9(1): 5178, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30518756

RESUMO

The development of new lymphatic vessels occurs in many cancerous and inflammatory diseases through the binding of VEGF-C to its receptors, VEGFR-2 and VEGFR-3. The regulation of VEGFR-2/VEGFR-3 heterodimerisation and its downstream signaling in lymphatic endothelial cells (LECs) remain poorly understood. Here, we identify the endocytic receptor, uPARAP, as a partner of VEGFR-2 and VEGFR-3 that regulates their heterodimerisation. Genetic ablation of uPARAP leads to hyperbranched lymphatic vasculatures in pathological conditions without affecting concomitant angiogenesis. In vitro, uPARAP controls LEC migration in response to VEGF-C but not VEGF-A or VEGF-CCys156Ser. uPARAP restricts VEGFR-2/VEGFR-3 heterodimerisation and subsequent VEGFR-2-mediated phosphorylation and inactivation of Crk-II adaptor. uPARAP promotes VEGFR-3 signaling through the Crk-II/JNK/paxillin/Rac1 pathway. Pharmacological Rac1 inhibition in uPARAP knockout mice restores the wild-type phenotype. In summary, our study identifies a molecular regulator of lymphangiogenesis, and uncovers novel molecular features of VEGFR-2/VEGFR-3 crosstalk and downstream signaling during VEGF-C-driven LEC sprouting in pathological conditions.


Assuntos
Linfangiogênese , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Dimerização , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Humanos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Receptores de Superfície Celular/genética , Transdução de Sinais , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética
8.
Sci Rep ; 7: 41494, 2017 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-28128294

RESUMO

Lymphangiogenesis, the formation of new lymphatic vessels, occurs in primary tumors and in draining lymph nodes leading to pre-metastatic niche formation. Reliable in vivo models are becoming instrumental for investigating alterations occurring in lymph nodes before tumor cell arrival. In this study, we demonstrate that B16F10 melanoma cell encapsulation in a biomaterial, and implantation in the mouse ear, prevents their rapid lymphatic spread observed when cells are directly injected in the ear. Vascular remodeling in lymph nodes was detected two weeks after sponge implantation, while their colonization by tumor cells occurred two weeks later. In this model, a huge lymphangiogenic response was induced in primary tumors and in pre-metastatic and metastatic lymph nodes. In control lymph nodes, lymphatic vessels were confined to the cortex. In contrast, an enlargement and expansion of lymphatic vessels towards paracortical and medullar areas occurred in pre-metastatic lymph nodes. We designed an original computerized-assisted quantification method to examine the lymphatic vessel structure and the spatial distribution. This new reliable and accurate model is suitable for in vivo studies of lymphangiogenesis, holds promise for unraveling the mechanisms underlying lymphatic metastases and pre-metastatic niche formation in lymph nodes, and will provide new tools for drug testing.


Assuntos
Bioensaio , Metástase Linfática/patologia , Vasos Linfáticos/patologia , Modelos Biológicos , Animais , Proliferação de Células , Gelatina , Processamento de Imagem Assistida por Computador , Implantes Experimentais , Injeções Intradérmicas , Linfonodos/patologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL
9.
Methods Mol Biol ; 1447: 331-49, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27514814

RESUMO

Therapeutic perspectives targeting angiogenesis in cancer stimulated an intense investigation of the mechanisms triggering and governing angiogenic processes. Several publications have highlighted the importance of typical dual-specificity phosphatases (DSPs) or MKPs in endothelial cells and their role in controlling different biological functions implicated in angiogenesis such as migration, proliferation, apoptosis, tubulogenesis, and cell adhesion. However, among atypical DSPs, the only one investigated in angiogenesis was DUSP3. We recently identified this DSP as a new key player in endothelial cells and angiogenesis. In this chapter we provide with detailed protocols and models used to investigate the role of DUSP3 in endothelial cells and angiogenesis. We start the chapter with an overview of the role of several DSPs in angiogenesis. We continue with providing a full description of a highly efficient transfection protocol to deplete DUSP3 using small interfering RNA (siRNA) in the primary human umbilical vein endothelial cells (HUVEC). We next describe the major assays used to investigate different processes involved in angiogenesis such as tube formation assay, proliferation assay and spheroids sprouting assay. We finish the chapter by validating our results in DUSP3-knockout mice using in vivo angiogenesis assays such as Matrigel plug and Lewis lung carcinoma cell subcutaneous xenograft model followed by anti-CD31 immunofluorescence and ex vivo aortic ring assay. All methods described can be adapted to other phosphatases and signaling molecules.


Assuntos
Fosfatase 3 de Especificidade Dupla/metabolismo , Células Endoteliais/citologia , Neovascularização Patológica , Neovascularização Fisiológica , Animais , Carcinoma Pulmonar de Lewis/metabolismo , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Fosfatase 3 de Especificidade Dupla/genética , Células Endoteliais/metabolismo , Imunofluorescência/métodos , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Transfecção/métodos
10.
PLoS One ; 9(5): e97019, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24806521

RESUMO

The endothelial cell spheroid assay provides a suitable in vitro model to study (lymph) angiogenesis and test pro- and anti-(lymph) angiogenic factors or drugs. Usually, the extent of cell invasion, observed through optical microscopy, is measured. The present study proposes the spatial distribution of migrated cells as a new descriptor of the (lymph) angiogenic response. The utility of this novel method rests with its capacity to locally characterise spheroid structure, allowing not only the investigation of single and collective cell invasion but also the evolution of the spheroid core itself. Moreover, the proposed method can be applied to 2D-projected spheroid images obtained by optical microscopy, as well as to 3D images acquired by confocal microscopy. To validate the proposed methodology, endothelial cell invasion was evaluated under different experimental conditions. The results were compared with widely used global parameters. The comparison shows that our method prevents local spheroid modifications from being overlooked and leading to the possible misinterpretation of results.


Assuntos
Rastreamento de Células/métodos , Células Endoteliais/ultraestrutura , Neovascularização Patológica/genética , Esferoides Celulares/ultraestrutura , Inibidores da Angiogênese/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Endoteliais/efeitos dos fármacos , Humanos , Imageamento Tridimensional , Microscopia Confocal , Neovascularização Patológica/tratamento farmacológico , Esferoides Celulares/efeitos dos fármacos , Telomerase/química , Telomerase/isolamento & purificação , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
PLoS One ; 9(9): e106976, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25222747

RESUMO

It is now well accepted that multipotent Bone-Marrow Mesenchymal Stem Cells (BM-MSC) contribute to cancer progression through several mechanisms including angiogenesis. However, their involvement during the lymphangiogenic process is poorly described. Using BM-MSC isolated from mice of two different backgrounds, we demonstrate a paracrine lymphangiogenic action of BM-MSC both in vivo and in vitro. Co-injection of BM-MSC and tumor cells in mice increased the in vivo tumor growth and intratumoral lymphatic vessel density. In addition, BM-MSC or their conditioned medium stimulated the recruitment of lymphatic vessels in vivo in an ear sponge assay, and ex vivo in the lymphatic ring assay (LRA). In vitro, MSC conditioned medium also increased the proliferation rate and the migration of both primary lymphatic endothelial cells (LEC) and an immortalized lymphatic endothelial cell line. Mechanistically, these pro-lymphangiogenic effects relied on the secretion of Vascular Endothelial Growth Factor (VEGF)-A by BM-MSC that activates VEGF Receptor (VEGFR)-2 pathway on LEC. Indeed, the trapping of VEGF-A in MSC conditioned medium by soluble VEGF Receptors (sVEGFR)-1, -2 or the inhibition of VEGFR-2 activity by a specific inhibitor (ZM 323881) both decreased LEC proliferation, migration and the phosphorylation of their main downstream target ERK1/2. This study provides direct unprecedented evidence for a paracrine lymphangiogenic action of BM-MSC via the production of VEGF-A which acts on LEC VEGFR-2.


Assuntos
Linfangiogênese , Células-Tronco Mesenquimais/fisiologia , Animais , Movimento Celular , Proliferação de Células , Meios de Cultivo Condicionados , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Feminino , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Oncotarget ; 5(7): 1856-68, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24732135

RESUMO

Beside a role in normal development/differentiation, high p63 immunoreactivity is also frequently observed in squamous cell carcinoma (SCC). Due to the complexity of the gene, the role of each p63 isotype in tumorigenesis is still confusing. Constitutively produced or induced in inflammatory conditions, human beta-defensins (HßDs) are cationic peptides involved in host defenses against bacteria, viruses and fungi. Here, we investigated both the role of p63 proteins in the regulation of HßDs and the implication of these antimicrobial peptides in tumor (lymph)angiogenesis. Thus, in contrast to TAp63 isotypes, we observed that ΔNp63 proteins (α, ß, γ) induce HßD1, 2 and 4 expression. Similar results were observed in cancer tissues and cell lines. We next demonstrated that ΔNp63-overexpressing SCC are associated with both a poor prognosis and a high tumor vascularisation and lymphangiogenesis. Moreover, we showed that HßDs exert a chemotactic activity for (lymphatic) endothelial cells in a CCR6-dependent manner. The ability of HßDs to enhance (lymph)angiogenesis in vivo was also evaluated. We observed that HßDs increase the vessel number and induce a significant increase in relative vascular area compared to negative control. Taken together, the results of this study suggest that ΔNp63-regulated HßD could promote tumor (lymph)angiogenesis in SCC microenvironment.


Assuntos
Carcinoma de Células Escamosas/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Linfangiogênese , Neovascularização Patológica/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Neoplasias do Colo do Útero/genética , beta-Defensinas/genética , Adulto , Idoso , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/química , Linhagem Celular Tumoral , Quimiotaxia , Células Endoteliais , Feminino , Genes Supressores de Tumor , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Neoplasias de Cabeça e Pescoço/química , Humanos , Queratinócitos , Masculino , Pessoa de Meia-Idade , Prognóstico , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Receptores CCR6/metabolismo , Taxa de Sobrevida , Fatores de Transcrição/análise , Fatores de Transcrição/metabolismo , Transfecção , Microambiente Tumoral , Proteínas Supressoras de Tumor/análise , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima , Neoplasias do Colo do Útero/irrigação sanguínea , Neoplasias do Colo do Útero/química , beta-Defensinas/análise , beta-Defensinas/metabolismo
13.
Invest Ophthalmol Vis Sci ; 54(5): 3082-93, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23580490

RESUMO

PURPOSE: To evaluate the antilymphangiogenic potential of multi-target tyrosine kinase inhibitor sunitinib in corneal neovascularization (NV). METHODS: Inflammatory corneal NV was induced by thermal cauterization applied in the central cornea of mice, to which sunitinib malate was daily administered by gavage or not. At days 6, 11, or 17 post cauterization, lymphatic and blood vessels, as well as inflammatory cells were immunostained and quantified in whole-mounted corneas. RT-PCRs were performed to evidence VEGF-A, VEGF-C, VEGF-D, placental growth factor (PlGF), and soluble vascular endothelial growth factor receptor (VEGFR)-1 and -2 (sVEGFR-1, sVEGFR-2) expressions. Macrophages were isolated from mice peritoneal cavity following thioglycollate injection to produce conditioned medium. The effects of sunitinib were evaluated in vitro in the aortic and lymphatic ring assays in the presence or not of macrophage conditioned medium. RESULTS: Sunitinib treatment drastically reduced pathologic corneal lymphangiogenesis and angiogenesis. Reduced F4/80+ cell infiltration was evidenced in sunitinib-treated mice and was associated to decreased VEGF-A (by 50%, P < 0.01) and VEGF-C (by 35%, P < 0.01) expressions, while VEGF-D and sVEGFR-2 expressions were not affected. In vitro, sunitinib dose-dependently inhibited aortic ring outgrowth, but failed to affect lymphangiogenesis in the lymphatic ring assay. However, macrophage conditioned medium-enhanced angiogenesis and lymphangiogenesis were both strongly counteracted by sunitinib treatment. Mechanistically, sunitinib blocked VEGFR-2 phosphorylation induced by VEGF-A released by macrophages. CONCLUSIONS: Sunitinib exerts antihemangiogenic and antilymphangiogenic effects in vivo by reducing F4/80+ cell recruitment and interacting with their released factors.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neovascularização da Córnea/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Indóis/uso terapêutico , Linfangiogênese/efeitos dos fármacos , Pirróis/uso terapêutico , Animais , Proliferação de Células/efeitos dos fármacos , Neovascularização da Córnea/genética , Neovascularização da Córnea/metabolismo , Modelos Animais de Doenças , Técnica Indireta de Fluorescência para Anticorpo , Glicoproteínas/metabolismo , Vasos Linfáticos/efeitos dos fármacos , Vasos Linfáticos/metabolismo , Macrófagos Peritoneais , Masculino , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Placentário , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas da Gravidez/genética , Proteínas Tirosina Quinases/antagonistas & inibidores , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sunitinibe , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA