Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Genes Dev ; 27(19): 2086-98, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24065739

RESUMO

Serine and cysteine cathepsin (Cts) proteases are an important class of intracellular and pericellular enzymes mediating multiple aspects of tumor development. Emblematic of these is CtsB, reported to play functionally significant roles during pancreatic islet and mammary carcinogenesis. CtsC, on the other hand, while up-regulated during pancreatic islet carcinogenesis, lacks functional significance in mediating neoplastic progression in that organ. Given that protein expression and enzymatic activity of both CtsB and CtsC are increased in numerous tumors, we sought to understand how tissue specificity might factor into their functional significance. Thus, whereas others have reported that CtsB regulates metastasis of mammary carcinomas, we found that development of squamous carcinomas occurs independently of CtsB. In contrast to these findings, our studies found no significant role for CtsC during mammary carcinogenesis but revealed squamous carcinogenesis to be functionally dependent on CtsC. In this context, dermal/stromal fibroblasts and bone marrow-derived cells expressed increased levels of enzymatically active CtsC that regulated the complexity of infiltrating immune cells in neoplastic skin, development of angiogenic vasculature, and overt squamous cell carcinoma growth. These studies highlight the important contribution of tissue/microenvironment context to solid tumor development and indicate that tissue specificity defines functional significance for these two members of the cysteine protease family.


Assuntos
Carcinogênese/genética , Carcinogênese/metabolismo , Carcinoma de Células Escamosas/fisiopatologia , Catepsina C/metabolismo , Neoplasias Cutâneas/fisiopatologia , Animais , Catepsina B/genética , Catepsina B/metabolismo , Catepsina C/genética , Linhagem Celular Tumoral , Quimases/metabolismo , Feminino , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Leucócitos/metabolismo , Neoplasias Mamárias Animais/fisiopatologia , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/genética , Elastase Pancreática/metabolismo
2.
Trends Immunol ; 33(3): 119-26, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22277903

RESUMO

Of the multiple unique stromal cell types common to solid tumors, tumor-associated macrophages (TAMs) are significant for fostering tumor progression. The protumor properties of TAMs derive from regulation of angiogenic programming, production of soluble mediators that support proliferation, survival and invasion of malignant cells, and direct and indirect suppression of cytotoxic T cell activity. These varied activities are dependent on the polarization state of TAMs that is regulated in part by local concentrations of cytokines and chemokines, as well as varied interactions of TAMs with normal and degraded components of the extracellular matrix. Targeting molecular pathways regulating TAM polarization holds great promise for anticancer therapy.


Assuntos
Macrófagos/imunologia , Neoplasias/imunologia , Microambiente Tumoral , Animais , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica
3.
Front Immunol ; 14: 1337333, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38313431

RESUMO

This review article will focus on subpopulations of fibroblasts that get reprogrammed by tumor cells into cancer-associated fibroblasts. Throughout this article, we will discuss the intricate interactions between fibroblasts, immune cells, and tumor cells. Unravelling complex intercellular crosstalk will pave the way for new insights into cellular mechanisms underlying the reprogramming of the local tumor immune microenvironment and propose novel immunotherapy strategies that might have potential in harnessing and modulating immune system responses.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias , Fibroblastos/patologia , Imunoterapia , Imunidade , Microambiente Tumoral
4.
Methods Mol Biol ; 539: 1-32, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19377975

RESUMO

Much progress has been made in understanding how matrix remodeling proteases, including metalloproteinases, serine proteases, and cysteine cathepsins, functionally contribute to cancer development. In addition to modulating extracellular matrix metabolism, proteases provide a significant protumor advantage to developing neoplasms through their ability to modulate bioavailability of growth and proangiogenic factors, regulation of bioactive chemokines and cytokines, and processing of cell-cell and cell-matrix adhesion molecules. Although some proteases directly regulate these events, it is now evident that some proteases indirectly contribute to cancer development by regulating posttranslational activation of latent zymogens that then directly impart regulatory information. Thus, many proteases act in a cascade-like manner and exert their functionality as part of a proteolytic pathway rather than simply functioning individually. Delineating the cascade of enzymatic activities contributing to overall proteolysis during carcinogenesis may identify rate-limiting steps or pathways that can be targeted with anti-cancer therapeutics. This chapter highlights recent insights into the complexity of roles played by pericellular and intracellular proteases by examining mechanistic studies as well as the roles of individual protease gene functions in various organ-specific mouse models of cancer development, with an emphasis on intersecting proteolytic activities that amplify programming of tissues to foster neoplastic development.


Assuntos
Neoplasias/enzimologia , Peptídeo Hidrolases/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Animais , Cisteína Endopeptidases/química , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteases/genética , Metaloproteases/metabolismo , Neoplasias/tratamento farmacológico , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/uso terapêutico , Serina Endopeptidases/análise , Serina Endopeptidases/metabolismo
5.
Anticancer Res ; 26(4B): 2805-20, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16886599

RESUMO

BACKGROUND: The goal of the present studies was to localize two proteins known to be involved in regulation of cell proliferation and survival in specific cell populations in normal SENCAR mouse skin and during multi-stage skin carcinogenesis. The proteins evaluated included activated Akt, as defined by phosphorylation of Akt at Serine-473 (pAkt) and mammalian target of rapamycin (pmTOR), defined by phosphorylation of mTOR at Serine-2448 (pmTOR). The cell populations examined included mouse keratinocyte stem cells (KSCs) within hair follicles and preneoplastic papilloma cells. MATERIALS AND METHODS: Immunochemical staining analysis as well as triple color immunofluorescence in combination with confocal microscopy were used to evaluate the presence of activated Akt and mammalian target of rapamycin (mTOR) in KSCs within the bulge niche of hair follicles, as identified by expression of the specific markers of mouse KSCs, CD34 and cytokeratin 15 (K15). Western blot analysis was used to examine CD34 and K15 protein levels in dorsal skin isolated from SENCAR mice during multi-stage skin carcinogenesis. RESULTS: CD34+/K15+ KSCs were located only in the outer root sheath (ORS) of a specific niche within hair follicles defined as "the bulge". The location of CD34+/K15+ KSCs remained restricted to the bulge region throughout the 22-week time-period examined during which pre-malignant papillomas developed and rapidly expanded. There was a significant decrease in K15 protein levels at 24 h and 15 weeks in dorsal skin treated with DMBA/TPA compared to CD34 protein levels. CD34+ cells within the numerous hair follicles in hyperplastic skin were found to undergo proliferation during the process of multi-stage skin carcinogenesis based on their staining with antibodies directed against proliferating cell nuclear antigen (PCNA). While pAkt was present within the bulge region of hair follicles, pmTOR was present in cells in the ORS of the bulge region as well as the upper infundibulum of hair follicles in dorsal skin treated with acetone. Within papillomas tissues isolated at 15 weeks following DMBA/TPA treatment, pAkt was localized to suprabasal cells with nominal staining of pAkt in the basal cell layer. There were fewer cells within the basal cell layer that contained pmTOR, in addition to the presence of pmTOR in suprabasal cells within papillomas. CONCLUSION: These results provide first time evidence for pAkt and pmTOR in CD34+/K15+ KSCs localized to the outer root sheath niche of the bulge region of mouse hair follicles. Taken together, the present observations suggest that pAkt and pmTOR may allow this cell population to evade terminal differentiation and to persist for long periods of time in their specific niche. Strategies that target pAkt and pmTOR may deplete both cells within the CD34+/K]5+ KSCs compartment, as well as impacting the survival of nonproliferating suprabasal cells within pre-malignant papillomas.


Assuntos
Antígenos CD34/biossíntese , Queratinócitos/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Cutâneas/metabolismo , Células-Tronco/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Ativação Enzimática , Feminino , Folículo Piloso/citologia , Folículo Piloso/metabolismo , Queratinócitos/patologia , Camundongos , Camundongos Endogâmicos SENCAR , Papiloma/metabolismo , Papiloma/patologia , Fosforilação , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/patologia , Células-Tronco/patologia , Serina-Treonina Quinases TOR
6.
Cancer Discov ; 6(3): 270-85, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26715645

RESUMO

UNLABELLED: Pancreas ductal adenocarcinoma (PDAC) has one of the worst 5-year survival rates of all solid tumors, and thus new treatment strategies are urgently needed. Here, we report that targeting Bruton tyrosine kinase (BTK), a key B-cell and macrophage kinase, restores T cell-dependent antitumor immune responses, thereby inhibiting PDAC growth and improving responsiveness to standard-of-care chemotherapy. We report that PDAC tumor growth depends on cross-talk between B cells and FcRγ(+) tumor-associated macrophages, resulting in T(H)2-type macrophage programming via BTK activation in a PI3Kγ-dependent manner. Treatment of PDAC-bearing mice with the BTK inhibitor PCI32765 (ibrutinib) or by PI3Kγ inhibition reprogrammed macrophages toward a T(H)1 phenotype that fostered CD8(+) T-cell cytotoxicity, and suppressed PDAC growth, indicating that BTK signaling mediates PDAC immunosuppression. These data indicate that pharmacologic inhibition of BTK in PDAC can reactivate adaptive immune responses, presenting a new therapeutic modality for this devastating tumor type. SIGNIFICANCE: We report that BTK regulates B-cell and macrophage-mediated T-cell suppression in pancreas adenocarcinomas. Inhibition of BTK with the FDA-approved inhibitor ibrutinib restores T cell-dependent antitumor immune responses to inhibit PDAC growth and improves responsiveness to chemotherapy, presenting a new therapeutic modality for pancreas cancer.


Assuntos
Comunicação Celular/imunologia , Sistema Imunitário/citologia , Sistema Imunitário/patologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Tirosina Quinase da Agamaglobulinemia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Humanos , Leucócitos/imunologia , Leucócitos/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias Pancreáticas/genética , Receptores de IgG/metabolismo , Transdução de Sinais
7.
Cancer Res ; 75(8): 1675-81, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25878147

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense stromal fibroinflammatory reaction that is a major obstacle to effective therapy. The desmoplastic stroma comprises many inflammatory cells, in particular mast cells as key components of the PDAC microenvironment, and such infiltration correlates with poor patient outcome. Indeed, it has been hypothesized that stromal ablation is critical to improve clinical response in patients with PDAC. Ibrutinib is a clinically approved Bruton's tyrosine kinase inhibitor that inhibits mast cells and tumor progression in a mouse model of ß-cell tumorigenesis. Here, we show that ibrutinib is highly effective at limiting the growth of PDAC in both transgenic mouse and patient-derived xenograft models of the disease. In these various experimental settings, ibrutinib effectively diminished fibrosis, extended survival, and improved the response to clinical standard-of-care therapy. Our results offer a preclinical rationale to immediately evaluate the clinical efficacy of ibrutinib in patients with PDAC.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Adenina/análogos & derivados , Adenocarcinoma/patologia , Animais , Antineoplásicos/farmacologia , Feminino , Fibrose/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias Pancreáticas/patologia , Piperidinas , Pirazóis/farmacologia , Pirimidinas/farmacologia , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Anticancer Res ; 24(5A): 2773-81, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15517884

RESUMO

The goal of the present study was to identify specific populations of cells that contain activated Akt-1, as determined by the presence ofphosphorylated Akt at serine 473 (p Akt), during development of skin tumors using a murine multi-stage carcinogenesis model. Nucleated papillomas cells as well as both epidermal and follicular keratinocytes in hyperplastic skin contained increased pAkt compared to skin treated only with acetone or 7, 12 dimethylbenz[a]anthracene (DMBA). Although the numbers of both mast cells and neutrophils were significantly increased in the stroma of papillomas (p<0.0005; p<0.0001, respectively), only mast cells contained pAkt. The amount of total Akt protein was similar regardless of time or treatment group examined. The present results suggest that activation of Akt-1 may provide specific populations of epidermal keratinocytes that develop into skin tumors with the ability to resist terminal differentiation and have enhanced proliferation during multi-stage skin carcinogenesis. In addition, mast cells which contain activated Akt-1 may persist within the stroma of papillomas during skin tumor development and progression through this signaling pathway, thereby contributing to a pro-oxidant and proangiogenic microenvironment.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Neoplasias Cutâneas/enzimologia , Pele/enzimologia , 9,10-Dimetil-1,2-benzantraceno , Acetona , Animais , Western Blotting , Carcinógenos , Ativação Enzimática , Feminino , Imunofluorescência , Imuno-Histoquímica , Queratinócitos/enzimologia , Queratinócitos/patologia , Mastócitos/enzimologia , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos SENCAR , Neutrófilos/enzimologia , Neutrófilos/patologia , Papiloma/induzido quimicamente , Papiloma/enzimologia , Papiloma/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Pele/efeitos dos fármacos , Pele/patologia , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Células Estromais/efeitos dos fármacos , Células Estromais/enzimologia , Acetato de Tetradecanoilforbol
9.
In Vivo ; 18(5): 525-42, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15523889

RESUMO

This review focuses on vascular endothelial growth factors (VEGF), a group of structurally-related proteins that serve as key growth factors for tumor-associated angiogenesis. Pathways induced by VEGF proteins that regulate biological functions of key cell types involved in tumor angiogenesis, including vascular endothelial cells, pericytes and tumor cells, are discussed. Strategies that are currently being developed and tested based on the emerging definitions of the roles of the multiple cell types involved in tumor vessel development, their selective production of VEGF-related proteins and other pro-angiogenic growth factors, their expression of associated receptors as well as identification of signal transduction pathways involved in VEGF-induced tumor survival and tumor-associated angiogenesis will be reviewed.


Assuntos
Progressão da Doença , Neoplasias/irrigação sanguínea , Neovascularização Patológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Humanos , Neoplasias/patologia , Neoplasias/fisiopatologia , Transdução de Sinais
10.
Cancer Cell ; 25(6): 809-821, 2014 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-24909985

RESUMO

B cells foster squamous cell carcinoma (SCC) development through deposition of immunoglobulin-containing immune complexes in premalignant tissue and Fcγ receptor-dependent activation of myeloid cells. Because human SCCs of the vulva and head and neck exhibited hallmarks of B cell infiltration, we examined B cell-deficient mice and found reduced support for SCC growth. Although ineffective as a single agent, treatment of mice bearing preexisting SCCs with B cell-depleting αCD20 monoclonal antibodies improved response to platinum- and Taxol-based chemotherapy. Improved chemoresponsiveness was dependent on altered chemokine expression by macrophages that promoted tumor infiltration of activated CD8(+) lymphocytes via CCR5-dependent mechanisms. These data reveal that B cells, and the downstream myeloid-based pathways they regulate, represent tractable targets for anticancer therapy in select tumors.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfócitos B/imunologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/imunologia , Macrófagos/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Células CHO , Carcinoma de Células Escamosas/patologia , Cricetulus , Feminino , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Neoplasias/patologia , Compostos Organoplatínicos/administração & dosagem , Paclitaxel/administração & dosagem , Fenótipo , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cytokine Growth Factor Rev ; 21(1): 3-10, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20005150

RESUMO

The classic view that the role of immune cells in cancer is primarily one of tumor rejection has been supplanted by a more complex view of leukocytes having both pro- and anti-tumor properties. This shift is due to the now well recognized capabilities of several myeloid cell types that foster pro-tumor programming of premalignant tissue, as well as the discovery that subsets of leukocytes also suppress development and effector functions of lymphocytes important for mediating anti-tumor immunity. In this review, we focus on the underappreciated role that T lymphocytes play in promoting tumor development. This includes, in addition to the role of T regulatory cells, a role for natural killer T cells and CD4(+) T helper cells in suppressing anti-tumor immunity and promoting cancer growth and metastasis.


Assuntos
Neoplasias/etiologia , Neoplasias/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Citocinas/imunologia , Humanos , Células Matadoras Naturais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Modelos Imunológicos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia
12.
Cancer Cell ; 17(2): 121-34, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-20138013

RESUMO

Chronically activated leukocytes recruited to premalignant tissues functionally contribute to cancer development; however, mechanisms underlying pro- versus anti-tumor programming of neoplastic tissues by immune cells remain obscure. Using the K14-HPV16 mouse model of squamous carcinogenesis, we report that B cells and humoral immunity foster cancer development by activating Fcgamma receptors (FcgammaRs) on resident and recruited myeloid cells. Stromal accumulation of autoantibodies in premalignant skin, through their interaction with activating FcgammaRs, regulate recruitment, composition, and bioeffector functions of leukocytes in neoplastic tissue, which in turn promote neoplastic progression and subsequent carcinoma development. These findings support a model in which B cells, humoral immunity, and activating FcgammaRs are required for establishing chronic inflammatory programs that promote de novo carcinogenesis.


Assuntos
Linfócitos B/imunologia , Carcinoma de Células Escamosas/imunologia , Neoplasias Epiteliais e Glandulares/imunologia , Receptores de IgG/fisiologia , Animais , Linfócitos B/metabolismo , Antígeno CD11b/metabolismo , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Imunidade Humoral/fisiologia , Mastócitos/imunologia , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias Epiteliais e Glandulares/irrigação sanguínea , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neovascularização Patológica , Receptores de IgG/metabolismo
13.
Dis Model Mech ; 3(5-6): 317-32, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20223936

RESUMO

Innate regulatory networks within organs maintain tissue homeostasis and facilitate rapid responses to damage. We identified a novel pathway regulating vessel stability in tissues that involves matrix metalloproteinase 14 (MMP14) and transforming growth factor beta 1 (TGFbeta(1)). Whereas plasma proteins rapidly extravasate out of vasculature in wild-type mice following acute damage, short-term treatment of mice in vivo with a broad-spectrum metalloproteinase inhibitor, neutralizing antibodies to TGFbeta(1), or an activin-like kinase 5 (ALK5) inhibitor significantly enhanced vessel leakage. By contrast, in a mouse model of age-related dermal fibrosis, where MMP14 activity and TGFbeta bioavailability are chronically elevated, or in mice that ectopically express TGFbeta in the epidermis, cutaneous vessels are resistant to acute leakage. Characteristic responses to tissue damage are reinstated if the fibrotic mice are pretreated with metalloproteinase inhibitors or TGFbeta signaling antagonists. Neoplastic tissues, however, are in a constant state of tissue damage and exhibit altered hemodynamics owing to hyperleaky angiogenic vasculature. In two distinct transgenic mouse tumor models, inhibition of ALK5 further enhanced vascular leakage into the interstitium and facilitated increased delivery of high molecular weight compounds into premalignant tissue and tumors. Taken together, these data define a central pathway involving MMP14 and TGFbeta that mediates vessel stability and vascular response to tissue injury. Antagonists of this pathway could be therapeutically exploited to improve the delivery of therapeutics or molecular contrast agents into tissues where chronic damage or neoplastic disease limits their efficient delivery.


Assuntos
Vasos Sanguíneos/enzimologia , Vasos Sanguíneos/patologia , Metaloproteinase 14 da Matriz/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Envelhecimento/patologia , Animais , Colágenos Fibrilares/metabolismo , Homeostase , Metaloproteinase 14 da Matriz/deficiência , Camundongos , Modelos Biológicos , Mostardeira , Óleos de Plantas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/patologia , Células Estromais/enzimologia , Células Estromais/patologia , Resistência Vascular
14.
Cell ; 129(1): 25-6, 2007 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-17418780

RESUMO

The development of solid tumors is regulated by dynamic interactions between evolving neoplastic cells and their microenvironment. Luo et al. (2007) recently demonstrated that tumor-infiltrating immune cells expressing RANKL induce activation and nuclear localization of IKKalpha in prostatic epithelial tumor cells. This leads to repression of maspin, a critical suppressor of metastasis, and thus commits malignant prostatic epithelial cells to a metastatic fate.


Assuntos
Quinase I-kappa B/metabolismo , Inflamação/metabolismo , Metástase Neoplásica , Neoplasias da Próstata/patologia , Animais , Células Epiteliais , Genes Supressores de Tumor , Humanos , Inflamação/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias da Próstata/metabolismo , Ligante RANK/metabolismo , Serpinas/genética , Serpinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa