Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Cell ; 40(9): 1027-1043.e9, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36099881

RESUMO

Programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1)-blockade immunotherapies have limited efficacy in the treatment of bladder cancer. Here, we show that NKG2A associates with improved survival and responsiveness to PD-L1 blockade immunotherapy in bladder tumors that have high abundance of CD8+ T cells. In bladder tumors, NKG2A is acquired on CD8+ T cells later than PD-1 as well as other well-established immune checkpoints. NKG2A+ PD-1+ CD8+ T cells diverge from classically defined exhausted T cells through their ability to react to human leukocyte antigen (HLA) class I-deficient tumors using T cell receptor (TCR)-independent innate-like mechanisms. HLA-ABC expression by bladder tumors is progressively diminished as disease progresses, framing the importance of targeting TCR-independent anti-tumor functions. Notably, NKG2A+ CD8+ T cells are inhibited when HLA-E is expressed by tumors and partly restored upon NKG2A blockade in an HLA-E-dependent manner. Overall, our study provides a framework for subsequent clinical trials combining NKG2A blockade with other T cell-targeted immunotherapies, where tumors express higher levels of HLA-E.


Assuntos
Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Neoplasias da Bexiga Urinária , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos , Antígenos de Histocompatibilidade Classe I , Humanos , Receptor de Morte Celular Programada 1 , Neoplasias da Bexiga Urinária/terapia , Antígenos HLA-E
2.
Nat Commun ; 12(1): 6889, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34824220

RESUMO

Inflammatory Breast Cancer (IBC) is a highly aggressive malignancy with distinct clinical and histopathological features whose molecular basis is unresolved. Here we describe a human IBC cell line, A3250, that recapitulates key IBC features in a mouse xenograft model, including skin erythema, diffuse tumor growth, dermal lymphatic invasion, and extensive metastases. A3250 cells express very high levels of the CCL2 chemokine and induce tumors enriched in macrophages. CCL2 knockdown leads to a striking reduction in macrophage densities, tumor proliferation, skin erythema, and metastasis. These results establish IBC-derived CCL2 as a key factor driving macrophage expansion, and indirectly tumor growth, with transcriptomic analysis demonstrating the activation of multiple inflammatory pathways. Finally, primary human IBCs exhibit macrophage infiltration and an enriched macrophage RNA signature. Thus, this human IBC model provides insight into the distinctive biology of IBC, and highlights potential therapeutic approaches to this deadly disease.


Assuntos
Quimiocina CCL2/metabolismo , Neoplasias Inflamatórias Mamárias/metabolismo , Neoplasias Inflamatórias Mamárias/patologia , Animais , Linhagem Celular Tumoral , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Inflamação , Neoplasias Inflamatórias Mamárias/genética , Neoplasias Inflamatórias Mamárias/imunologia , Camundongos , Camundongos SCID , Células Mieloides/metabolismo , Metástase Neoplásica , Receptores CCR2/metabolismo , Transplante Heterólogo , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/patologia
3.
Nat Commun ; 12(1): 4447, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34290243

RESUMO

Tryptophan catabolism is a major metabolic pathway utilized by several professional and non-professional antigen presenting cells to maintain immunological tolerance. Here we report that 3-hydroxy-L-kynurenamine (3-HKA) is a biogenic amine produced via an alternative pathway of tryptophan metabolism. In vitro, 3-HKA has an anti-inflammatory profile by inhibiting the IFN-γ mediated STAT1/NF-κΒ pathway in both mouse and human dendritic cells (DCs) with a consequent decrease in the release of pro-inflammatory chemokines and cytokines, most notably TNF, IL-6, and IL12p70. 3-HKA has protective effects in an experimental mouse model of psoriasis by decreasing skin thickness, erythema, scaling and fissuring, reducing TNF, IL-1ß, IFN-γ, and IL-17 production, and inhibiting generation of effector CD8+ T cells. Similarly, in a mouse model of nephrotoxic nephritis, besides reducing inflammatory cytokines, 3-HKA improves proteinuria and serum urea nitrogen, overall ameliorating immune-mediated glomerulonephritis and renal dysfunction. Overall, we propose that this biogenic amine is a crucial component of tryptophan-mediated immune tolerance.


Assuntos
Aminas Biogênicas/farmacologia , Imunomodulação/efeitos dos fármacos , Cinurenina/análogos & derivados , Animais , Aminas Biogênicas/metabolismo , Aminas Biogênicas/uso terapêutico , Linhagem Celular Tumoral , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Células Endoteliais , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Inflamação , Interferon gama/farmacologia , Cinurenina/metabolismo , Cinurenina/farmacologia , Cinurenina/uso terapêutico , Camundongos , NF-kappa B/metabolismo , Nefrite/tratamento farmacológico , Nefrite/imunologia , Psoríase/tratamento farmacológico , Psoríase/imunologia , Triptofano/metabolismo
4.
Integr Biol (Camb) ; 13(1): 1-16, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33443535

RESUMO

Tumor emboli-aggregates of tumor cells within vessels-pose a clinical challenge as they are associated with increased metastasis and tumor recurrence. When growing within a vessel, tumor emboli are subject to a unique mechanical constraint provided by the tubular geometry of the vessel. Current models of tumor emboli use unconstrained multicellular tumor spheroids, which neglect this mechanical interplay. Here, we modeled a lymphatic vessel as a 200 µm-diameter channel in either a stiff or soft, bioinert agarose matrix to create a vessel-like constraint model (VLCM), and we modeled colon or breast cancer tumor emboli with aggregates of HCT116 or SUM149PT cells, respectively. The stiff matrix VLCM constrained the tumor emboli to the cylindrical channel, which led to continuous growth of the emboli, in contrast to the growth rate reduction that unconstrained spheroids exhibit. Emboli morphology in the soft matrix VLCM, however, was dependent on the magnitude of mechanical mismatch between the matrix and the cell aggregates. In general, when the elastic modulus of the matrix of the VLCM was greater than the emboli (EVLCM/Eemb > 1), the emboli were constrained to grow within the channel, and when the elastic modulus of the matrix was less than the emboli (0 < EVLCM/Eemb < 1), the emboli bulged into the matrix. Due to a large difference in myosin II expression between the cell lines, we hypothesized that tumor cell aggregate stiffness is an indicator of cellular force-generating capability. Inhibitors of myosin-related force generation decreased the elastic modulus and/or increased the stress relaxation of the tumor cell aggregates, effectively increasing the mechanical mismatch. The increased mechanical mismatch after drug treatment was correlated with increased confinement of tumor emboli growth along the channel, which may translate to increased tumor burden due to the increased tumor volume within the diffusion distance of nutrients and oxygen.


Assuntos
Neoplasias da Mama , Vasos Linfáticos , Células Neoplásicas Circulantes , Feminino , Humanos , Esferoides Celulares
5.
Front Oncol ; 10: 571100, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33194666

RESUMO

In primary breast tumors, cancer cells hematogenously disseminate through doorways in the vasculature composed of three-cell complexes (known as Tumor MicroEnvironment of Metastasis) comprising a perivascular macrophage, a tumor cell overexpressing the actin-regulatory protein Mammalian Enabled (Mena), and an endothelial cell, all in direct physical contact. It has been previously shown that once tumor cells establish lymph node metastases in patients, TMEM doorways form in the metastatic tumor cell nests. However, it has not been established if such lymph node-TMEM doorways actively transit tumor cells into the peripheral circulation and on to tertiary sites. To address this question in this short report, we used a mouse model of lymph node metastasis to demonstrate that TMEM doorways: (1) exist in tumor-positive lymph nodes of mice, (2) are restricted to the blood vascular endothelium, (3) serve as a mechanism for further dissemination to peripheral sites such as to the lungs, and (4) their activity can be abrogated by a pharmaceutical intervention. Our data suggest that cancer cell dissemination via TMEM doorways is a common mechanism of breast cancer cell dissemination to distant sites and thus the pharmacological targeting of TMEM may be necessary, even after resection of the primary tumor, to suppress cancer cell dissemination.

6.
Methods Mol Biol ; 1846: 279-290, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30242766

RESUMO

Studies of lymphangiogenesis and lymphatic endothelial biology in vitro require pure cultures of lymphatic endothelial cells and 3D vascular constructs, which closely resemble native human lymphatic vasculature. We describe a method for the isolation of human dermal microvascular lymphatic endothelial cells and generation of a 3D lymphatic capillary network. The lymphatic vascular construct is generated by coculturing primary lymphatic endothelial cells and fibroblasts in their native matrix, without the use of synthetic scaffolds or exogenous factors. The tissue is stable over many weeks and accurately recapitulates features of human dermal lymphatic microvasculature.


Assuntos
Células Endoteliais/metabolismo , Vasos Linfáticos/citologia , Vasos Linfáticos/metabolismo , Pele/metabolismo , Biomarcadores , Separação Celular/métodos , Células Cultivadas , Endotélio Linfático , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Imageamento Tridimensional/métodos , Imuno-Histoquímica , Pele/irrigação sanguínea , Fluxo de Trabalho
7.
Nat Protoc ; 12(5): 1077-1088, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28448484

RESUMO

This protocol describes a unique in vitro method for the generation of a 3D human lymphatic network within native connective tissue devoid of any exogenous material such as scaffolds or growth factors. In this five-stage protocol, human lymphatic endothelial cells (LECs) cocultured with dermal fibroblasts spontaneously organize into a stable 3D lymphatic capillary network. Stage 1 involves the isolation of primary fibroblasts and LECs from human skin. Fibroblasts are then cultured to produce connective tissue rich in extracellular matrix (stage 2), onto which LECs are seeded to form a network (stage 3). After stacking of tissue layers and tissue maturation at the air-liquid interface (stage 4), the 3D construct containing the lymphatic microvascular network can be analyzed by microscopy (stage 5). Lymphatic vasculature generated by this approach exhibits the major cellular and ultrastructural features of native in vivo human dermal lymphatic microvasculature and is stable over many weeks. The protocol for generating a 3D construct takes 6 weeks to complete, and it requires experience in cell culture techniques. The system described here offers a unique opportunity to study the mechanisms underlying lymphatic vessel formation, remodeling and function in a human cell context.


Assuntos
Linfangiogênese/fisiologia , Microvasos , Técnicas de Cultura de Órgãos/métodos , Técnicas de Cocultura/métodos , Células Endoteliais/fisiologia , Fibroblastos/fisiologia , Humanos
8.
Biomaterials ; 78: 129-39, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26694987

RESUMO

Regeneration of lymphatic vessels is important for treatment of various disorders of lymphatic system and for restoration of lymphatic function after surgery. We have developed a method for generating a human 3D lymphatic vascular construct. In this system, human lymphatic endothelial cells, co-cultured with fibroblasts, spontaneously organized into a stable 3D lymphatic capillary network without the use of any exogenous factors. In vitro-generated lymphatic capillaries exhibited the major molecular and ultra-structural features of native, human lymphatic microvasculature: branches in the three dimensions, wide lumen, blind ends, overlapping borders, adherens and tight junctions, anchoring filaments, lack of mural cells, and poorly developed basement membrane. Furthermore, we show that fibroblast-derived VEGF-C and HGF cooperate in the formation of lymphatic vasculature by activating ERK1/2 signaling, and demonstrate distinct functions of HGF/c-Met and VEGF-C/VEGFR-3 in lymphangiogenesis. This lymphatic vascular construct is expected to facilitate studies of lymphangiogenesis in vitro and it holds promise as a strategy for regeneration of lymphatic vessels and treatment of lymphatic disorders in various conditions.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Vasos Linfáticos/anatomia & histologia , Fator C de Crescimento do Endotélio Vascular/metabolismo , Humanos , Técnicas In Vitro
9.
Microvasc Res ; 95: 46-52, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25026412

RESUMO

In cancer, lymphatic vasculature has been traditionally viewed only as a transportation system for metastatic cells. It has now become clear that lymphatics perform many additional functions which could influence cancer progression. Lymphangiogenesis, induced at the primary tumor site and at distant sites, potently augments metastasis. Lymphatic endothelial cells (LECs) control tumor cell entry and exit from the lymphatic vessels. LECs also control immune cell traffic and directly modulate adaptive immune responses. This review highlights advances in our understanding of the mechanisms by which lymphatic vessels, and in particular lymphatic endothelium, impact metastasis.


Assuntos
Células Endoteliais/patologia , Vasos Linfáticos/patologia , Neoplasias/patologia , Imunidade Adaptativa , Animais , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Linfangiogênese , Metástase Linfática , Vasos Linfáticos/imunologia , Vasos Linfáticos/metabolismo , Vasos Linfáticos/fisiopatologia , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Prognóstico
10.
ACS Nano ; 7(11): 10362-70, 2013 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-24134041

RESUMO

In the current study we show the dissociation and tumor accumulation dynamics of dual-labeled near-infrared quantum dot core self-assembled lipidic nanoparticles (SALNPs) in a mouse model upon intravenous administration. Using advanced in vivo fluorescence energy transfer imaging techniques, we observed swift exchange with plasma protein components in the blood and progressive SALNP dissociation and subsequent trafficking of individual SALNP components following tumor accumulation. Our results suggest that upon intravenous administration SALNPs quickly transform, which may affect their functionality. The presented technology provides a modular in vivo tool to visualize SALNP behavior in real time and may contribute to improving the therapeutic outcome or molecular imaging signature of SALNPs.


Assuntos
Nanopartículas/análise , Administração Intravenosa , Animais , Linhagem Celular Tumoral , Feminino , Transferência Ressonante de Energia de Fluorescência/métodos , Humanos , Cinética , Lipídeos/química , Camundongos , Micelas , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Imagem Molecular , Nanopartículas/química , Nanotecnologia , Transplante de Neoplasias , Óptica e Fotônica , Pontos Quânticos
11.
J Exp Med ; 210(8): 1509-28, 2013 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-23878309

RESUMO

Lymphatic vessels are thought to contribute to metastasis primarily by serving as a transportation system. It is widely believed that tumor cells enter lymph nodes passively by the flow of lymph. We demonstrate that lymph node lymphatic sinuses control tumor cell entry into the lymph node, which requires active tumor cell migration. In human and mouse tissues, CCL1 protein is detected in lymph node lymphatic sinuses but not in the peripheral lymphatics. CCR8, the receptor for CCL1, is strongly expressed by human malignant melanoma. Tumor cell migration to lymphatic endothelial cells (LECs) in vitro is inhibited by blocking CCR8 or CCL1, and recombinant CCL1 promotes migration of CCR8(+) tumor cells. The proinflammatory mediators TNF, IL-1ß, and LPS increase CCL1 production by LECs and tumor cell migration to LECs. In a mouse model, blocking CCR8 with the soluble antagonist or knockdown with shRNA significantly decreased lymph node metastasis. Notably, inhibition of CCR8 led to the arrest of tumor cells in the collecting lymphatic vessels at the junction with the lymph node subcapsular sinus. These data identify a novel function for CCL1-CCR8 in metastasis and lymph node LECs as a critical checkpoint for the entry of metastases into the lymph nodes.


Assuntos
Quimiocina CCL1/metabolismo , Linfonodos/metabolismo , Linfonodos/patologia , Neoplasias/imunologia , Neoplasias/patologia , Animais , Linhagem Celular Tumoral , Movimento Celular/imunologia , Fatores Quimiotáticos/metabolismo , Quimiotaxia/imunologia , Citocinas/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Endotélio Linfático/efeitos dos fármacos , Endotélio Linfático/imunologia , Endotélio Linfático/metabolismo , Humanos , Mediadores da Inflamação/farmacologia , Linfonodos/imunologia , Metástase Linfática , Vasos Linfáticos/imunologia , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Melanoma/imunologia , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Microscopia de Fluorescência por Excitação Multifotônica , Receptores CCR8/antagonistas & inibidores , Receptores CCR8/metabolismo , Imagem com Lapso de Tempo
13.
Cancer Res ; 70(5): 1814-24, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20179201

RESUMO

The lymphatic system is an important pathway for tumor dissemination to the lymph nodes, but to which extent it contributes to the formation of distant metastases remains unknown. We report that induction of lymphangiogenesis by vascular endothelial growth factor-C (VEGF-C) at the secondary site, in the lung, facilitates expansion of already disseminated cancer cells throughout the lung tissue. By using orthotopic spontaneous metastasis models in nude mice, we show that VEGF-C expression by tumor cells altered the pattern of pulmonary metastases from nodular to diffuse and facilitated disease progression. Metastases expressing VEGF-C were tightly associated with the airways, in contrast to the control cells that were scattered in the lung parenchyma, throughout the alveolar region. VEGF-C induced lung lymphangiogenesis and promoted intralymphatic spread of metastases in the lung and formation of tumor emboli in the pulmonary arteries. This pattern of metastasis corresponds to lymphangitic carcinomatosis metastatic phenotype in human cancer patients, an extremely aggressive pattern of pulmonary metastases. In accordance, pulmonary breast cancer metastases from patients which were classified as lymphangitic carcinomatosis showed high levels of VEGF-C expression in cancer cells. These data show that VEGF-C promotes late steps of the metastatic process and identify the VEGF-C/VEGF receptor-3 pathway as the target not only for prevention of metastases, but also for treatment of established metastatic disease.


Assuntos
Carcinoma/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Linfangite/patologia , Fator C de Crescimento do Endotélio Vascular/biossíntese , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma/irrigação sanguínea , Carcinoma/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/patologia , Linfonodos/metabolismo , Linfonodos/patologia , Linfangiogênese , Linfangite/metabolismo , Metástase Linfática , Camundongos , Camundongos Nus
14.
Am J Pathol ; 176(2): 981-94, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20042679

RESUMO

Vascular endothelial growth factor (VEGF), which is a key regulator of angiogenesis, often induces formation of immature vessels with increased permeability and reduced vessel functionality. Here, we demonstrate that de novo expression of murine (m)VEGF-164 induces malignant and invasive tumor growth of HaCaT keratinocytes. However, the mVEGF-164-induced tumors are ulcerated with a disorganized epithelium that is interrupted by lacunae with limited basement membrane and endothelial cell coverage. Vessel maturation is strongly impaired. Tumor and vessel micromorphology are markedly improved by the combined expression of human platelet-derived growth factor (hPDGF)-B and mVEGF-164. Although tumor size and malignancy are comparable with either mVEGF-164 alone or combined human PDGF-B and mVEGF-164 expression, combined hPDGF-B and mVEGF-164 expression leads to a more solid and compact tumor tissue with a mature functional tumor vasculature and a higher microvessel density, as demonstrated histologically and by dynamic contrast-enhanced magnetic resonance imaging. Treatment of the hPDGF-B- and mVEGF-164-expressing tumors with imatinib mesylate to block PDGF-B signaling reverses this effect. In addition, tumor cell invasion of mVEGF-164 transfectants and mVEGF-164 plus hPDGF-B transfectants in vivo is associated with a marked induction of tumor-derived matrix metalloproteinase-1 and stromal matrix metalloproteinase-9 and -13, as was confirmed in three-dimensional organotypic co-cultures with fibroblasts in vitro. These data clearly demonstrate the need for a concerted action of different growth factors in the establishment of solid tumors with functional vasculature and emphasize the need for a multifactorial therapy.


Assuntos
Carcinoma de Células Escamosas/patologia , Neovascularização Patológica/fisiopatologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Neoplasias Cutâneas/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Benzamidas , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/ultraestrutura , Proliferação de Células , Células Cultivadas , Humanos , Mesilato de Imatinib , Camundongos , Camundongos Nus , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-sis/genética , Pirimidinas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/ultraestrutura , Transfecção , Transplante Heterólogo , Carga Tumoral/genética
16.
J Immunol ; 183(3): 1767-79, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19587009

RESUMO

The lymphatic system is essential for the generation of immune responses by facilitating immune cell trafficking to lymph nodes. Dendritic cells (DCs), the most potent APCs, exit tissues via lymphatic vessels, but the mechanisms of interaction between DCs and the lymphatic endothelium and the potential implications of these interactions for immune responses are poorly understood. In this study, we demonstrate that lymphatic endothelial cells (LECs) modulate the maturation and function of DCs. Direct contact of human monocyte-derived DCs with an inflamed, TNF-alpha-stimulated lymphatic endothelium reduced expression of the costimulatory molecule CD86 by DCs and suppressed the ability of DCs to induce T cell proliferation. These effects were dependent on adhesive interactions between DCs and LECs that were mediated by the binding of Mac-1 on DCs to ICAM-1 on LECs. Importantly, the suppressive effects of the lymphatic endothelium on DCs were observed only in the absence of pathogen-derived signals. In vivo, DCs that migrated to the draining lymph nodes upon inflammatory stimuli, but in the absence of a pathogen, showed increased levels of CD86 expression in ICAM-1-deficient mice. Together, these data demonstrate a direct role of LECs in the modulation of immune response and suggest a function of the lymphatic endothelium in preventing undesired immune reactions in inflammatory conditions.


Assuntos
Células Dendríticas/patologia , Endotélio Linfático/fisiopatologia , Molécula 1 de Adesão Intercelular/imunologia , Antígeno de Macrófago 1/imunologia , Animais , Antígeno B7-2/análise , Adesão Celular/imunologia , Diferenciação Celular , Técnicas de Cocultura , Endotélio Linfático/patologia , Humanos , Imunidade , Inflamação , Molécula 1 de Adesão Intercelular/metabolismo , Antígeno de Macrófago 1/metabolismo , Camundongos , Ligação Proteica
17.
Ann N Y Acad Sci ; 1131: 235-41, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18519976

RESUMO

Most cancerous lesions metastasize through the lymphatic system and the status of regional lymph nodes is the most important indicator of a patient's prognosis. The extent of lymph node involvement with cancer is also an important parameter used for determining treatment options. Although the importance of the lymphatic system for metastasis has been well recognized, traditionally, the lymphatic vessels have not been considered actively involved in the metastatic process. Recent evidence, however, indicates that the activation of the lymphatic system is an important factor in tumor progression to metastasis. Tumor lymphangiogenesis has been associated with increased propensity for metastasis, and lymphatic vessel density has emerged as another promising prognostic indicator. More recently, lymphangiogenesis in the sentinel lymph nodes has been shown to contribute to malignant progression. In addition to its role as a transport system for tumor cells, the lymphatic system may also be more actively involved in metastases by directly facilitating tumor cell recruitment into the lymphatic vessels. This review highlights recent advances in our understanding of the mechanisms by which lymphatic vessels participate in metastasis.


Assuntos
Vasos Linfáticos/fisiopatologia , Neoplasias/fisiopatologia , Humanos , Linfangiogênese/fisiologia , Metástase Linfática/patologia , Metástase Linfática/prevenção & controle , Vasos Linfáticos/patologia , Neoplasias/metabolismo , Neoplasias/patologia , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Biópsia de Linfonodo Sentinela , Fatores de Crescimento do Endotélio Vascular/metabolismo
18.
J Clin Invest ; 117(11): 3369-82, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17948123

RESUMO

The Notch family of cell surface receptors and its ligands are highly conserved proteins that regulate cell fate determination, including those involved in mammalian vascular development. We report that Notch induces VEGFR-3 expression in vitro in human endothelial cells and in vivo in mice. In vitro, Notch in complex with the DNA-binding protein CBF-1/suppressor of hairless/Lag1 (CSL) bound the VEGFR-3 promoter and transactivated VEGFR-3 specifically in endothelial cells. Through induction of VEGFR-3, Notch increased endothelial cell responsiveness to VEGF-C, promoting endothelial cell survival and morphological changes. In vivo, VEGFR-3 was upregulated in endothelial cells with active Notch signaling. Mice heterozygous for null alleles of both Notch1 and VEGFR-3 had significantly reduced viability and displayed midgestational vascular patterning defects analogous to Notch1 nullizygous embryos. We found that Notch1 and Notch4 were expressed in normal and tumor lymphatic endothelial cells and that Notch1 was activated in lymphatic endothelium of invasive mammary micropapillary carcinomas. These results demonstrate that Notch1 and VEGFR-3 interact genetically, that Notch directly induces VEGFR-3 in blood endothelial cells to regulate vascular development, and that Notch may function in tumor lymphangiogenesis.


Assuntos
Células Endoteliais/metabolismo , Receptores Notch/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Forma Celular , Sobrevivência Celular , Células Cultivadas , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Células Endoteliais/citologia , Feminino , Regulação da Expressão Gênica , Humanos , Camundongos , Receptores Notch/genética , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética
19.
Proc Natl Acad Sci U S A ; 104(12): 5026-31, 2007 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-17360402

RESUMO

Infiltration of lymphocytes into the thyroid gland and formation of lymph node-like structures is a hallmark of Hashimoto's thyroiditis. Here we demonstrate that lymphatic vessels are present within these infiltrates. Mice overexpressing the chemokine CCL21 in the thyroid (TGCCL21 mice) developed similar lymphoid infiltrates and lymphatic vessels. TGCCL21 mice lacking mature T and B cells (RAGTGCCL21 mice) did not have cellular infiltrates or increased number of lymphatic vessels compared with controls. Transfer of CD3(+)CD4(+) T cells into RAGTGCCL21 mice promoted the development of LYVE-1(+)podoplanin(+)Prox-1(+) vessels in the thyroid. Genetic deletion of lymphotoxin beta receptor or lymphotoxin alpha abrogated development of lymphatic vessels in the inflamed areas in the thyroid but did not affect development of neighboring lymphatics. These results define a model for the study of inflammatory lymphangiogenesis in the thyroid and implicate lymphotoxin beta receptor signaling in this process.


Assuntos
Linfangiogênese/imunologia , Receptor beta de Linfotoxina/metabolismo , Transdução de Sinais , Glândula Tireoide/patologia , Animais , Linfócitos T CD4-Positivos/imunologia , Separação Celular , Quimiocina CCL21 , Quimiocinas CC/metabolismo , Doença de Hashimoto/imunologia , Doença de Hashimoto/patologia , Humanos , Inflamação , Vasos Linfáticos/imunologia , Vasos Linfáticos/patologia , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , Receptor beta de Linfotoxina/deficiência , Linfotoxina-alfa/deficiência , Camundongos , Camundongos Transgênicos , Glândula Tireoide/imunologia
20.
Am J Pathol ; 169(5): 1767-83, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17071599

RESUMO

Platelet-derived growth factor (PDGF) stimulates tumor growth and progression by affecting tumor and stromal cells. In the HaCaT skin carcinogenesis model, transfection of immortal nontumorigenic and PDGF-receptor-negative HaCaT keratinocytes with PDGF-B induced formation of benign tumors. Here, we present potential mechanisms underlying this tumorigenic conversion. In vivo, persistent PDGF-B expression induced enhanced tumor cell proliferation but only transiently stimulated stromal cell proliferation and angiogenesis. In vitro and in vivo studies identified fibroblasts as PDGF target cells essential for mediating transient angiogenesis and persistent epithelial hyperproliferation. In fibroblast cultures, long-term PDGF-BB treatment caused an initial up-regulation of vascular endothelial growth factor (VEGF)-A, followed by a drastic VEGF down-regulation and myofibroblast differentiation. Accordingly, in HaCaT/PDGF-B transplants, initially enhanced VEGF expression by stromal fibroblasts was subsequently reduced, followed by down-regulation of angiogenesis, myofibroblast accumulation, and vessel maturation. The PDGF-induced, persistently increased expression of the hepatocyte growth factor by fibroblasts in vitro and in vivo was most probably responsible for enhanced epithelial cell proliferation and benign tumor formation. Thus, by paracrine stimulation of the stroma, PDGF-BB induced epithelial hyperproliferation, thereby promoting tumorigenicity, whereas the time-limited activation of the stroma followed by stromal maturation provides a possible explanation for the benign tumor phenotype.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Substâncias de Crescimento/farmacologia , Neoplasias/patologia , Fenótipo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Células Estromais/efeitos dos fármacos , Actinas/metabolismo , Becaplermina , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica , Células Cultivadas , Endostatinas/metabolismo , Células Epiteliais/citologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Queratinócitos/citologia , Queratinócitos/patologia , Mesoderma/citologia , Neovascularização Fisiológica/efeitos dos fármacos , Comunicação Parácrina/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Células Estromais/citologia , Células Estromais/patologia , Fatores de Tempo , Transfecção , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...