Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Nat Cell Biol ; 9(7): 838-46, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17572668

RESUMO

Human immunodeficiency virus (HIV)-1 infection requires envelope (Env) glycoprotein gp120-induced clustering of CD4 and coreceptors (CCR5 or CXCR4) on the cell surface; this enables Env gp41 activation and formation of a complex that mediates fusion between Env-containing and target-cell membranes. Kinetic studies show that viral receptors are actively transported to the Env-receptor interface in a process that depends on plasma membrane composition and the actin cytoskeleton. The mechanisms by which HIV-1 induces F-actin rearrangement in the target cell remain largely unknown. Here, we show that CD4 and the coreceptors interact with the actin-binding protein filamin-A, whose binding to HIV-1 receptors regulates their clustering on the cell surface. We found that gp120 binding to cell receptors induces transient cofilin-phosphorylation inactivation through a RhoA-ROCK-dependent mechanism. Blockade of filamin-A interaction with CD4 and/or coreceptors inhibits gp120-induced RhoA activation and cofilin inactivation. Our results thus identify filamin-A as an adaptor protein that links HIV-1 receptors to the actin cytoskeleton remodelling machinery, which may facilitate virus infection.


Assuntos
Actinas/fisiologia , Antígenos CD4/metabolismo , Proteínas Contráteis/fisiologia , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/fisiologia , Proteínas dos Microfilamentos/fisiologia , Fatores de Despolimerização de Actina/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Filaminas , HIV-1/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
2.
Breast Cancer Res ; 15(4): R54, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23826634

RESUMO

INTRODUCTION: Dysregulated NOTCH receptor activity has been implicated in breast cancer but the mechanisms by which NOTCH contributes to transformation are not yet clear, as it has context-dependent effects on the properties of transformed cells. METHODS: We have used various in vitro and in vivo carcinogenic models to analyze the impact of Notch signaling in the onset and progression of breast tumors. RESULTS: We found that ectopic expression of the Notch1 intracellular domain (N1ICD) in MCF-7 breast adenocarcinoma cell line caused reduction and delocalization of E-CADHERIN levels and increased migratory and invasive abilities. Notch inhibition in the invasive breast cancer cell line MDA-MB-231 resulted in increased E-CADHERIN expression and a parallel reduction in their invasive capacity. The growth of subcutaneous xenografts produced with MCF-7 cells was boosted after N1ICD induction, in a cell autonomous manner. In vivo Notch1 activation in the mammary gland using the MMTV-Cre driver caused the formation of papillary tumors that showed increased Hes1 and Hey1 expression and delocalized E-cadherin staining. CONCLUSIONS: These results confirm NOTCH1 as a signal triggering epithelial-mesenchymal transition in epithelial cancer cells, which may have implications in tumor dissemination, metastasis and proliferation in vivo. The identification of specific factors interacting with NOTCH signaling could thus be relevant to fully understanding the role of NOTCH in breast neoplasia.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Receptores Notch/metabolismo , Animais , Neoplasias da Mama/genética , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células , Modelos Animais de Doenças , Regulação para Baixo , Células Epiteliais/metabolismo , Feminino , Expressão Gênica , Xenoenxertos , Humanos , Células MCF-7 , Glândulas Mamárias Humanas/metabolismo , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica , Domínios e Motivos de Interação entre Proteínas/genética , Receptor Notch1/química , Receptor Notch1/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores Notch/antagonistas & inibidores , Receptores Notch/química , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral
3.
Cancers (Basel) ; 14(5)2022 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-35267534

RESUMO

The balance between laminin isoforms containing the α5 or the α4 chain in the endothelial basement membrane determines the site of leukocyte diapedesis under inflammatory conditions. Extracellular superoxide dismutase (SOD3) induces laminin α4 expression in tumor blood vessels, which is associated with enhanced intratumor T cell infiltration in primary human cancers. We show now that SOD3 overexpression in neoplastic and endothelial cells (ECs) reduces laminin α5 in tumor blood vessels. SOD3 represses the laminin α5 gene (LAMA5), but LAMA5 expression is not changed in SOD1-overexpressing cells. Transcriptomic analyses revealed SOD3 overexpression to change the transcription of 1682 genes in ECs, with the canonical and non-canonical NF-κB pathways as the major SOD3 targets. Indeed, SOD3 reduced the transcription of well-known NF-κB target genes as well as NF-κB-driven promoter activity in ECs stimulated with tumor necrosis factor (TNF)-α, an NF-κB signaling inducer. SOD3 inhibited the phosphorylation and degradation of IκBα (nuclear factor of the kappa light polypeptide gene enhancer in B-cells inhibitor alpha), an NF-κB inhibitor. Finally, TNF-α was found to be a transcriptional activator of LAMA5 but not of LAMA4; LAMA5 induction was prevented by SOD3. In conclusion, SOD3 is a major regulator of laminin balance in the basement membrane of tumor ECs, with potential implications for immune cell infiltration into tumors.

4.
Ann Intern Med ; 153(5): 281-8, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20820037

RESUMO

BACKGROUND: Hepatic encephalopathy is a major complication of cirrhosis and is associated with a poor prognosis. OBJECTIVE: To identify mutations in the gene sequence for glutaminase in humans that could be responsible for the development of hepatic encephalopathy in patients with cirrhosis. DESIGN: Cohort study. SETTING: Outpatient clinics in 6 Spanish hospitals. PATIENTS: 109 consecutive patients with cirrhosis in the estimation cohort, 177 patients in the validation cohort, and 107 healthy control participants. MEASUREMENTS: Patients were followed every 3 or 6 months until the development of hepatic encephalopathy or liver transplantation, death, or the end of the study. RESULTS: The genetic analyses showed that glutaminase TACC and CACC haplotypes were linked to the risk for overt hepatic encephalopathy. Mutation scanning of the glutaminase gene identified a section in the promoter region where base pairs were repeated (a microsatellite). Over a mean follow-up of 29.6 months, hepatic encephalopathy occurred in 28 patients (25.7%) in the estimation cohort. Multivariable Cox models were used to determine the following independent predictors: Child-Turcotte-Pugh stage (hazard ratio [HR], 1.6 [95% CI, 1.29 to 1.98]; P = 0.001), minimal hepatic encephalopathy (HR, 3.17 [CI, 1.42 to 7.09]; P = 0.006), and having 2 long alleles of the microsatellite (HR, 3.12 [CI, 1.39 to 7.02]; P = 0.006). The association between 2 long alleles of the microsatellite and overt hepatic encephalopathy was confirmed in a validation cohort (HR, 2.1 [CI, 1.17 to 3.79]; P = 0.012). Functional studies showed higher luciferase activity in cells transfected with the long form of the microsatellite, which suggests that the long microsatellite enhances glutaminase transcriptional activity. LIMITATION: Other genes and allelic variants might be involved in the clinical expression of hepatic encephalopathy. CONCLUSION: This study identifies a genetic factor that is associated with development of hepatic encephalopathy in patients with cirrhosis. PRIMARY FUNDING SOURCE: Instituto de Salud Carlos III, Spanish Ministry of Health.


Assuntos
Glutaminase/genética , Encefalopatia Hepática/genética , Cirrose Hepática/enzimologia , Cirrose Hepática/genética , Mutação , Regiões Promotoras Genéticas/genética , Idoso , Feminino , Encefalopatia Hepática/etiologia , Humanos , Cirrose Hepática/complicações , Masculino , Repetições de Microssatélites/genética , Pessoa de Meia-Idade , Fatores de Risco
5.
Mol Cancer ; 9: 196, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20649976

RESUMO

BACKGROUND: ErbB2-positive breast cancer is characterized by highly aggressive phenotypes and reduced responsiveness to standard therapies. Although specific ErbB2-targeted therapies have been designed, only a small percentage of patients respond to these treatments and most of them eventually relapse. The existence of this population of particularly aggressive and non-responding or relapsing patients urges the search for novel therapies. The purpose of this study was to determine whether cannabinoids might constitute a new therapeutic tool for the treatment of ErbB2-positive breast tumors. We analyzed their antitumor potential in a well established and clinically relevant model of ErbB2-driven metastatic breast cancer: the MMTV-neu mouse. We also analyzed the expression of cannabinoid targets in a series of 87 human breast tumors. RESULTS: Our results show that both Delta9-tetrahydrocannabinol, the most abundant and potent cannabinoid in marijuana, and JWH-133, a non-psychotropic CB2 receptor-selective agonist, reduce tumor growth, tumor number, and the amount/severity of lung metastases in MMTV-neu mice. Histological analyses of the tumors revealed that cannabinoids inhibit cancer cell proliferation, induce cancer cell apoptosis, and impair tumor angiogenesis. Cannabinoid antitumoral action relies, at least partially, on the inhibition of the pro-tumorigenic Akt pathway. We also found that 91% of ErbB2-positive tumors express the non-psychotropic cannabinoid receptor CB2. CONCLUSIONS: Taken together, these results provide a strong preclinical evidence for the use of cannabinoid-based therapies for the management of ErbB2-positive breast cancer.


Assuntos
Neoplasias da Mama/patologia , Canabinoides/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Receptor ErbB-2/fisiologia , Neoplasias da Mama/metabolismo , Progressão da Doença , Regulação para Baixo , Feminino , Humanos , Metástase Neoplásica , Receptor CB2 de Canabinoide/metabolismo
6.
J Exp Med ; 200(4): 541-7, 2004 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-15314078

RESUMO

Human immunodeficiency virus (HIV)-1 infectivity requires actin-dependent clustering of host lipid raft-associated receptors, a process that might be linked to Rho guanosine triphosphatase (GTPase) activation. Rho GTPase activity can be negatively regulated by statins, a family of drugs used to treat hypercholesterolemia in man. Statins mediate inhibition of Rho GTPases by impeding prenylation of small G proteins through blockade of 3-hydroxy-3-methylglutaryl coenzyme A reductase. We show that statins decreased viral load and increased CD4+ cell counts in acute infection models and in chronically HIV-1-infected patients. Viral entry and exit was reduced in statin-treated cells, and inhibition was blocked by the addition of l-mevalonate or of geranylgeranylpyrophosphate, but not by cholesterol. Cell treatment with a geranylgeranyl transferase inhibitor, but not a farnesyl transferase inhibitor, specifically inhibited entry of HIV-1-pseudotyped viruses. Statins blocked Rho-A activation induced by HIV-1 binding to target cells, and expression of the dominant negative mutant RhoN19 inhibited HIV-1 envelope fusion with target cell membranes, reducing cell infection rates. We suggest that statins have direct anti-HIV-1 effects by targeting Rho.


Assuntos
Síndrome da Imunodeficiência Adquirida/metabolismo , Citoesqueleto/metabolismo , Regulação para Baixo/efeitos dos fármacos , HIV-1/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/metabolismo , Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Animais , Contagem de Linfócito CD4 , Células Cultivadas , Colesterol/sangue , Modelos Animais de Doenças , Eletroforese em Gel de Poliacrilamida , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Ácido Mevalônico , Camundongos , Camundongos SCID , Fosfatos de Poli-Isoprenil , Testes de Precipitina , RNA/metabolismo
7.
J Exp Med ; 196(3): 293-301, 2002 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12163558

RESUMO

Human immunodeficiency virus (HIV)-1 infection depends on multiple lateral interactions between the viral envelope and host cell receptors. Previous studies have suggested that these interactions are possible because HIV-1 receptors CD4, CXCR4, and CCR5 partition in cholesterol-enriched membrane raft domains. We generated CD4 partitioning mutants by substituting or deleting CD4 transmembrane and cytoplasmic domains and the CD4 ectodomain was unaltered. We report that all CD4 mutants that retain raft partitioning mediate HIV-1 entry and CD4-induced Lck activation independently of their transmembrane and cytoplasmic domains. Conversely, CD4 ectodomain targeting to a nonraft membrane fraction results in a CD4 receptor with severely diminished capacity to mediate Lck activation or HIV-1 entry, although this mutant binds gp120 as well as CD4wt. In addition, the nonraft CD4 mutant inhibits HIV-1 X4 and R5 entry in a CD4(+) cell line. These results not only indicate that HIV-1 exploits host membrane raft domains as cell entry sites, but also suggest new strategies for preventing HIV-1 infection.


Assuntos
Síndrome da Imunodeficiência Adquirida/terapia , Antígenos CD4/química , HIV-1/fisiologia , Microdomínios da Membrana/química , Sequência de Aminoácidos , Antígenos CD4/fisiologia , Linhagem Celular , Ativação Enzimática , Proteína gp120 do Envelope de HIV/fisiologia , Humanos , Lipoproteínas LDL/farmacologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Dados de Sequência Molecular , Receptores CXCR4/fisiologia
8.
J Exp Med ; 198(9): 1381-9, 2003 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-14597737

RESUMO

Chemokines are implicated in tumor pathogenesis, although it is unclear whether they affect human cancer progression positively or negatively. We found that activation of the chemokine receptor CCR5 regulates p53 transcriptional activity in breast cancer cells through pertussis toxin-, JAK2-, and p38 mitogen-activated protein kinase-dependent mechanisms. CCR5 blockade significantly enhanced proliferation of xenografts from tumor cells bearing wild-type p53, but did not affect proliferation of tumor xenografts bearing a p53 mutation. In parallel, data obtained in a primary breast cancer clinical series showed that disease-free survival was shorter in individuals bearing the CCR5Delta32 allele than in CCR5 wild-type patients, but only for those whose tumors expressed wild-type p53. These findings suggest that CCR5 activity influences human breast cancer progression in a p53-dependent manner.


Assuntos
Neoplasias da Mama/patologia , Receptores CCR5/genética , Proteína Supressora de Tumor p53/metabolismo , Neoplasias da Mama/metabolismo , Divisão Celular , Progressão da Doença , Humanos , Receptores CCR5/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
9.
J Immunol ; 181(5): 3524-34, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18714025

RESUMO

The statins, a group of inhibitors of the 3-hydroxy-3-methylglutaryl coenzyme A reductase, are reported to influence a variety of immune system activities through 3-hydroxy-3-methylglutaryl coenzyme A reductase-dependent and -independent mechanisms. How statin treatment regulates immune system function in vivo nonetheless remains to be fully defined. We analyzed the immunomodulatory effects of lovastatin in a Candida albicans-induced delayed-type hypersensitivity reaction in mice. In this model, lovastatin administration reduced the acute inflammatory response elicited by C. albicans challenge. This anti-inflammatory activity of lovastatin was associated with a shift from a Th1 to a Th2 immune response, as well as an increase in the percentage of regulatory T cells at the inflammation site and in the regional draining lymph node. The lovastatin-induced increase in regulatory T cells in the inflamed skin was dependent on expression of CCL1, a chemokine that is locally up-regulated by statin administration. The anti-inflammatory effect of lovastatin was abrogated in CCL1-deficient mice. These results suggest that local regulation of chemokine expression may be an important process in statin-induced modulation of the immune system.


Assuntos
Quimiocina CCL1/genética , Quimiotaxia de Leucócito/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Candida albicans/imunologia , Quimiotaxia de Leucócito/imunologia , Hipersensibilidade/microbiologia , Hipersensibilidade/patologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Lovastatina/administração & dosagem , Lovastatina/farmacologia , Camundongos , Camundongos Knockout , Subpopulações de Linfócitos T/efeitos dos fármacos , Linfócitos T Reguladores/fisiologia , Células Th2/imunologia , Regulação para Cima/efeitos dos fármacos
10.
Oncoimmunology ; 9(1): 1794163, 2020 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-32934887

RESUMO

The conversion of a non-T cell-inflamed into a T cell-inflamed tumor microenvironment (TME) is a key to sensitizing tumors to T-cell-based immunotherapies. Recent data show that the extracellular superoxide dismutase (SOD3) alters endothelial basement membrane (EC-BM) composition, providing permissive signals that enhance tumor infiltration by effector T cells. Abbreviations: AJ, adherens junction; EC, endothelial cell; EC-BM, endothelial basement membrane; HIF, hypoxia-inducible factor; ICAM-1, intercellular adhesion molecule-1; LAMA4, laminin-α4; SOD3, superoxide dismutase-3; TME, tumor microenvironment; VCAM-1, vascular cell adhesion molecule-1; VEGF, vascular-endothelial growth factor.


Assuntos
Laminina , Neoplasias , Endotélio Vascular , Humanos , Superóxido Dismutase/genética , Linfócitos T , Microambiente Tumoral
11.
Front Immunol ; 11: 579552, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33250894

RESUMO

Tumor-infiltrating lymphocytes (TILs) are major players in the immune-mediated control of cancer and the response to immunotherapy. In primary cancers, however, TILs are commonly absent, suggesting T-cell entry into the tumor microenvironment (TME) to be selectively restricted. Blood and lymph vessels are the first barriers that circulating T-cells must cross to reach the tumor parenchyma. Certainly, the crossing of the endothelial cell (EC) basement membrane (EC-BM)-an extracellular matrix underlying EC-is a limiting step in T-cell diapedesis. This review highlights new data suggesting the antioxidant enzyme superoxide dismutase-3 (SOD3) to be a regulator of EC-BM composition in the tumor vasculature. In the EC, SOD3 induces vascular normalization and endows the EC-BM with the capacity for the extravasation of effector T-cells into the TME, which it achieves via the WNT signaling pathway. However, when activated in tumor cells, this same pathway is reported to exclude TILs. SOD3 also regulates TIL density in primary human colorectal cancers (CRC), thus affecting the relapse rate and patient survival.


Assuntos
Membrana Basal/patologia , Neoplasias Colorretais/metabolismo , Células Endoteliais/patologia , Linfócitos do Interstício Tumoral/imunologia , Superóxido Dismutase/metabolismo , Linfócitos T/imunologia , Animais , Movimento Celular , Neoplasias Colorretais/patologia , Espaço Extracelular , Humanos , Microambiente Tumoral , Via de Sinalização Wnt
12.
J Immunother Cancer ; 8(1)2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32591431

RESUMO

BACKGROUND: Tumor-infiltrating lymphocytes (TILs), mainly CD8+ cytotoxic T lymphocytes (CTL), are linked to immune-mediated control of human cancers and response to immunotherapy. Tumors have nonetheless developed specific mechanisms that selectively restrict T cell entry into the tumor microenvironment. The extracellular superoxide dismutase (SOD3) is an anti-oxidant enzyme usually downregulated in tumors. We hypothesize that upregulation of SOD3 in the tumor microenvironment might be a mechanism to boost T cell infiltration by normalizing the tumor-associated endothelium. RESULTS: Here we show that SOD3 overexpression in endothelial cells increased in vitro transmigration of naïve and activated CD4+ and CD8+ T cells, but not of myeloid cells. Perivascular expression of SOD3 also specifically increased CD4+ and CD8+ effector T cell infiltration into tumors and improved the effectiveness of adoptively transferred tumor-specific CD8+ T cells. SOD3-induced enhanced transmigration in vitro and tumor infiltration in vivo were not associated to upregulation of T cell chemokines such as CXCL9 or CXCL10, nor to changes in the levels of endothelial adhesion receptors such as intercellular adhesion molecule-1 (ICAM-1) or vascular cell adhesion molecule-1 (VCAM-1). Instead, SOD3 enhanced T cell infiltration via HIF-2α-dependent induction of specific WNT ligands in endothelial cells; this led to WNT signaling pathway activation in the endothelium, FOXM1 stabilization, and transcriptional induction of laminin-α4 (LAMA4), an endothelial basement membrane component permissive for T cell infiltration. In patients with stage II colorectal cancer, SOD3 was associated with increased CD8+ TIL density and disease-free survival. SOD3 expression was also linked to a T cell-inflamed gene signature using the COAD cohort from The Cancer Genome Atlas program. CONCLUSION: Our findings suggest that SOD3-induced upregulation of LAMA4 in endothelial cells boosts selective tumor infiltration by T lymphocytes, thus transforming immunologically "cold" into "hot" tumors. High SOD3 levels are associated with human colon cancer infiltration by CD8+ T cells, with potential consequences for the clinical outcome of these patients. Our results also uncover a cell type-specific, distinct activity of the WNT pathway for the regulation of T cell infiltration into tumors.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Endoteliais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias/imunologia , Superóxido Dismutase/metabolismo , Superóxido Dismutase/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Feminino , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais , Superóxido Dismutase/genética , Células Tumorais Cultivadas , Microambiente Tumoral
13.
J Cell Biol ; 157(2): 277-89, 2002 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11956229

RESUMO

Cell signaling does not occur randomly over the cell surface, but is integrated within cholesterol-enriched membrane domains, termed rafts. By targeting SHP-2 to raft domains or to a non-raft plasma membrane fraction, we studied the functional role of rafts in signaling. Serum-depleted, nonattached cells expressing the raft SHP-2 form, but not non-raft SHP-2, display signaling events resembling those observed after fibronectin attachment, such as beta1 integrin clustering, 397Y-FAK phosphorylation, and ERK activation, and also increases Rho-GTP levels. Expression of the dominant negative N19Rho abrogates raft-SHP-2-induced signaling, suggesting that Rho activation is a downstream event in SHP-2 signaling. Expression of a catalytic inactive SHP-2 mutant abrogates the adhesion-induced feedback inhibition of Rho activity, suggesting that SHP-2 contributes to adhesion-induced suppression of Rho activity. Because raft recruitment of SHP-2 occurs physiologically after cell attachment, these results provide a mechanism by which SHP-2 may influence cell adhesion and migration by spatially regulating Rho activity.


Assuntos
Integrinas/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo , Western Blotting , Adesão Celular , Linhagem Celular , Membrana Celular/metabolismo , Tamanho Celular , Clonagem Molecular , Ativação Enzimática , Fibronectinas/metabolismo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Guanosina Trifosfato/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Microdomínios da Membrana/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Quinases/metabolismo , Fatores de Tempo , Transfecção , Proteínas rac de Ligação ao GTP/metabolismo
14.
J Cell Biol ; 164(5): 759-68, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14981096

RESUMO

Spatially restricted activation of signaling molecules governs critical aspects of cell migration; the mechanism by which this is achieved nonetheless remains unknown. Using time-lapse confocal microscopy, we analyzed dynamic redistribution of lipid rafts in chemoattractant-stimulated leukocytes expressing glycosyl phosphatidylinositol-anchored green fluorescent protein (GFP-GPI). Chemoattractants induced persistent GFP-GPI redistribution to the leading edge raft (L raft) and uropod rafts of Jurkat, HL60, and dimethyl sulfoxide-differentiated HL60 cells in a pertussis toxin-sensitive, actin-dependent manner. A transmembrane, nonraft GFP protein was distributed homogeneously in moving cells. A GFP-CCR5 chimera, which partitions in L rafts, accumulated at the leading edge, and CCR5 redistribution coincided with recruitment and activation of phosphatidylinositol-3 kinase gamma in L rafts in polarized, moving cells. Membrane cholesterol depletion impeded raft redistribution and asymmetric recruitment of PI3K to the cell side facing the chemoattractant source. This is the first direct evidence that lipid rafts order spatial signaling in moving mammalian cells, by concentrating the gradient sensing machinery at the leading edge.


Assuntos
Quimiotaxia/fisiologia , Leucócitos/metabolismo , Microdomínios da Membrana/metabolismo , Transdução de Sinais/fisiologia , Actinas/metabolismo , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Fatores Quimiotáticos/metabolismo , Ativação Enzimática , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Humanos , Leucócitos/citologia , Microscopia de Vídeo , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores CCR5/genética , Receptores CCR5/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
15.
Exp Biol Med (Maywood) ; 233(7): 860-73, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18445772

RESUMO

Pancreatic adenocarcinoma is characterized by desmoplasia, local invasion, and metastasis. These features are regulated in part by MMP9 and SPARC. To explore the interaction of SPARC and MMP9 in cancer, we first established orthotopic pancreatic tumors in SPARC-null and wild-type mice with the murine pancreatic adenocarcinoma cell line, PAN02. MMP9 expression was higher in tumors from wild-type compared to SPARC-null mice. Coincident with lower MMP9 expression, tumors grown in SPARC-null mice were significantly larger, had decreased ECM deposition and reduced microvessel density compared to wild-type controls. In addition, metastasis was enhanced in the absence of host SPARC. Therefore, we next analyzed the orthotopic tumor growth of PAN02 cells transduced with MMP9 or a control empty vector. Forced expression of MMP9 by the PAN02 cells resulted in larger tumors in both wild-type and SPARC-null animals compared to empty vector controls and further diminished ECM deposition. Importantly, forced expression of MMP9 within the tumor reversed the decrease in angiogenesis and abrogated the metastatic potential displayed by control tumors grown in SPARC-null mice. Finally, contrary to the in vivo results, MMP9 increased cell migration in vitro, which was blocked by the addition of SPARC. These results suggest that SPARC and MMP9 interact to regulate many stages of tumor progression including ECM deposition, angiogenesis and metastasis.


Assuntos
Adenocarcinoma/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Patológica/metabolismo , Osteonectina/metabolismo , Neoplasias Pancreáticas/metabolismo , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Regulação Neoplásica da Expressão Gênica , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Knockout , Metástase Neoplásica/patologia , Metástase Neoplásica/fisiopatologia , Osteonectina/genética , Neoplasias Pancreáticas/patologia
16.
Nat Commun ; 9(1): 575, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29422508

RESUMO

One drawback of chemotherapy is poor drug delivery to tumor cells, due in part to hyperpermeability of the tumor vasculature. Extracellular superoxide dismutase (SOD3) is an antioxidant enzyme usually repressed in the tumor milieu. Here we show that specific SOD3 re-expression in tumor-associated endothelial cells (ECs) increases doxorubicin (Doxo) delivery into and chemotherapeutic effect on tumors. Enhanced SOD3 activity fostered perivascular nitric oxide accumulation and reduced vessel leakage by inducing vascular endothelial cadherin (VEC) transcription. SOD3 reduced HIF prolyl hydroxylase domain protein activity, which increased hypoxia-inducible factor-2α (HIF-2α) stability and enhanced its binding to a specific VEC promoter region. EC-specific HIF-2α ablation prevented both the SOD3-mediated increase in VEC transcription and the enhanced Doxo effect. SOD3, VEC, and HIF-2α levels correlated positively in primary colorectal cancers, which suggests a similar interconnection of these proteins in human malignancy.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Doxorrubicina/administração & dosagem , Células Endoteliais/metabolismo , Neoplasias/tratamento farmacológico , Superóxido Dismutase/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Antineoplásicos/administração & dosagem , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Caderinas/genética , Caderinas/metabolismo , Dioxigenases/genética , Dioxigenases/metabolismo , Tratamento Farmacológico , Células Endoteliais/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/genética , Neoplasias/metabolismo , Estabilidade Proteica , Superóxido Dismutase/genética
17.
J Clin Invest ; 110(12): 1831-8, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12488433

RESUMO

Hepatocellular carcinoma is strongly associated with chronic infection by the hepatitis B virus (HBV) and has poor prognosis due to intrahepatic metastasis. HBx is often the only HBV protein detected in hepatic tumor cells; however, its contribution to tumor invasion and metastasis has not been established so far. In this work, we show that HBx enhances tumor cell invasion, both in vivo and in vitro. The increased invasive capacity induced by HBx is mediated by an upregulation of membrane-type 1 matrix metalloproteinase (MT1-MMP) expression, which in turn activates matrix metalloproteinase-2. Induction of both MT1-MMP expression and cell invasion by HBx is dependent on cyclooxygenase-2 (COX-2) activity. In addition, HBx upregulates the expression of COX-2, which is mediated by the transcriptional activation of the COX-2 gene promoter in a nuclear factor of activated T cell-dependent (NF-AT-dependent) manner. These results demonstrate the ability of HBx to promote tumor cell invasion by a mechanism involving the upregulation of MT1-MMP and COX-2 and provide new insights into the mechanism of action of this viral protein and its involvement in tumor metastasis and recurrence of hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular/patologia , Isoenzimas/metabolismo , Neoplasias Hepáticas/patologia , Metaloendopeptidases/metabolismo , Invasividade Neoplásica , Prostaglandina-Endoperóxido Sintases/metabolismo , Transativadores/metabolismo , Animais , Anti-Inflamatórios não Esteroides/metabolismo , Carcinoma Hepatocelular/enzimologia , Embrião de Galinha , Meios de Cultura Livres de Soro , Ciclo-Oxigenase 2 , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Antígenos da Hepatite B/metabolismo , Humanos , Isoenzimas/genética , Neoplasias Hepáticas/enzimologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinases da Matriz Associadas à Membrana , Proteínas de Membrana , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/genética , Prognóstico , Regiões Promotoras Genéticas , Prostaglandina-Endoperóxido Sintases/genética , Ativação Transcricional , Células Tumorais Cultivadas , Regulação para Cima/fisiologia , Proteínas Virais Reguladoras e Acessórias
18.
Clin Exp Metastasis ; 19(4): 313-8, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12090471

RESUMO

Tumor cells acquire the ability to enter blood vessels surrounding the primary tumor, endowing them with the capacity to disseminate and become established in distant sites, originating a metastasis. Determination of the intravasation ability of tumor cells is thus important, as it can be correlated with their potential malignancy. To analyze the intravasation phenotype of human tumor cells in vivo, we performed chick embryo chorioallantoic membrane (CAM) assays. Cells were inoculated on the CAM of 9-day-old chick embryos and the membrane at the opposite side of the egg was recovered after 48 h incubation. To measure intravasation ability, we calculated the amount of human DNA in each CAM sample by real-time PCR of Alu sequences and SYBR Green 1 fluorescence detection. This analysis showed a detection limit of 1 human cell per 10(5) total cells, and we were able to distinguish between tumor cells of distinct invasive capacity. This assay has several advantages over current methods to measure intravasation ability, including the elimination of post-PCR analysis, sensitivity and easy scale-up of sample numbers.


Assuntos
DNA de Neoplasias/análise , Invasividade Neoplásica , Compostos Orgânicos , Reação em Cadeia da Polimerase/métodos , Adenocarcinoma/patologia , Alantoide/patologia , Elementos Alu , Animais , Benzotiazóis , Neoplasias da Mama/patologia , Embrião de Galinha , Córion/patologia , Sistemas Computacionais , Diaminas , Etídio/análise , Fibroblastos , Corantes Fluorescentes/análise , Humanos , Fenótipo , Reação em Cadeia da Polimerase/instrumentação , Quinolinas , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/patologia
19.
Oncotarget ; 4(12): 2288-301, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24317954

RESUMO

Beyond their ability to inhibit cholesterol biosynthesis, the statins have pleiotropic effects that include anti-inflammatory and immunomodulatory activities. Statins could have clinical utility, alone or in combination with other chemotherapeutics, in the treatment of cancer. The mechanisms that underlie the anti-tumor activity of the statins are nonetheless poorly defined. No studies have analyzed how they alter the tumor-associated leukocyte infiltrate, a central factor that influences tumor stroma and cancer evolution. Here we used HER2/neu transgenic (Tg-neu) mice to analyze the effect of lovastatin (Lov) on the inflammatory reaction of spontaneous mammary tumors. Lov treatment of tumor-bearing Tg-neu mice did not alter growth of established tumors, but significantly reduced the number of new oncogenic lesions in these mice. Moreover, Lov inhibited the growth of newly implanted Tg-neu tumors in immunocompetent but not in immunodeficient mice. We found that Lov enhanced tumor infiltration by effector T cells, and reduced the number of immunosuppressive and pro-angiogenic M2-like tumor-associated macrophages (TAM). Concomitantly, the drug improved the structure and function of the tumor vasculature, measured as enhanced tumor oxygenation and penetration of cytotoxic drugs. Microarray analysis identified a Lov-elicited genetic program in Tg-neu tumors that might explain these effects; we observed Lov-induced downregulation of placental growth factor, which triggers aberrant angiogenesis and M2-like TAM polarization. Our results identify a role for lovastatin in the shaping and re-education of the inflammatory infiltrate in tumors, with functional consequences in angiogenesis and antitumor immunity.


Assuntos
Anticolesterolemiantes/farmacologia , Lovastatina/farmacologia , Macrófagos/efeitos dos fármacos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/imunologia , Linfócitos T/efeitos dos fármacos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Polaridade Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Feminino , Lovastatina/administração & dosagem , Macrófagos/imunologia , Macrófagos/patologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Linfócitos T/imunologia , Linfócitos T/metabolismo
20.
Cancer Res ; 73(14): 4461-73, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23720051

RESUMO

Chemokines are relevant molecules in shaping the tumor microenvironment, although their contributions to tumorigenesis are not fully understood. We studied the influence of the chemokine CX3CL1/fractalkine in de novo breast cancer formation using HER2/neu transgenic mice. CX3CL1 expression was downmodulated in HER2/neu tumors, yet, paradoxically, adenovirus-mediated CX3CL1 expression in the tumor milieu enhanced mammary tumor numbers in a dose-dependent manner. Increased tumor multiplicity was not a consequence of CX3CL1-induced metastatic dissemination of the primary tumor, although CX3CL1 induced epithelial-to-mesenchymal transition in breast cancer cells in vitro. Instead, CX3CL1 triggered cell proliferation by induction of ErbB receptors through the proteolytic shedding of an ErbB ligand. This effect was important insofar as mammary tumorigenesis was delayed and tumor multiplicity was reduced by genetic deletion of CX3CL1 in HER2/neu mice, but not in polyoma middle T-antigen oncomice. Our findings support the conclusion that CX3CL1 acts as a positive modifier of breast cancer in concert with ErbB receptors.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Ativação Transcricional , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Linfonodos/metabolismo , Linfonodos/patologia , Metástase Linfática , Células MCF-7 , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Animais , Neoplasias Mamárias Experimentais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA