Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Immunity ; 40(6): 949-60, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24909887

RESUMEN

Despite development of new antiviral drugs, viral infections are still a major health problem. The most potent antiviral defense mechanism is the innate production of type I interferon (IFN-I), which not only limits virus replication but also promotes antiviral T cell immunity through mechanisms, which remain insufficiently studied. Using the murine lymphocytic choriomeningitis virus model system, we show here that IFN-I signaling on T cells prevented their rapid elimination in vivo. Microarray analyses uncovered that IFN-I triggered the expression of selected inhibitory NK-cell-receptor ligands. Consequently, T cell immunity of IFN-I receptor (IFNAR)-deficient T cells could be restored by NK cell depletion or in NK-cell-deficient hosts (Nfil3(-/-)). The elimination of Ifnar1(-/-) T cells was dependent on NK-cell-mediated perforin expression. In summary, we identified IFN-I as a key player regulating the protection of T cells against regulatory NK cell function.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Interferón Tipo I/inmunología , Células Asesinas Naturales/inmunología , Coriomeningitis Linfocítica/inmunología , Receptor de Interferón alfa y beta/inmunología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Células Cultivadas , Inmunidad Innata , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Perforina/biosíntesis , Receptor de Interferón alfa y beta/genética , Transducción de Señal/inmunología , Replicación Viral/inmunología
2.
J Immunol ; 195(10): 4650-9, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26459352

RESUMEN

Type I IFNs (IFN-I) are cytokines that can mediate both immune suppression and activation. Dendritic cells (DC) are significant producers of IFN-I, and depending on the context (nature of Ag, duration of exposure to Ag), DC-derived IFN-I can have varying effects on CD8(+) T cell responses. In this study, we report that in the context of a CD8(+) T cell response to a self-Ag, DC-intrinsic expression of IFN regulatory factor 3 is required to induce optimal proliferation and migration of autoreactive CD8(+) T cells, ultimately determining their ability to infiltrate a target tissue (pancreas), and the development of glucose intolerance in rat insulin promoter-glycoprotein (RIP-GP) mice. Moreover, we show that signals through the lymphotoxin-ß receptor (LTßR) in DC are also required for the proliferation of autoreactive CD8(+) T cells, the upregulation of VLA4/LFA1 on activated CD8(+) T cells, and their subsequent infiltration into the pancreas both in vitro and in vivo. Importantly, the defects in autoreactive CD8(+) T cell proliferation, accumulation of CD8(+) T cells in the pancreas, and consequent glucose intolerance observed in the context of priming by LTßR(-/-) DC could be rescued by exogenous addition of IFN-I. Collectively, our data demonstrate that the LTßR/IFN-I axis is essential for programming of CD8(+) T cells to mediate immunopathology in a self-tissue. A further understanding of the IFN-I/LTßR axis will provide valuable therapeutic insights for treatment of CD8(+) T cell-mediated autoimmune diseases.


Asunto(s)
Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón Tipo I/inmunología , Receptor beta de Linfotoxina/inmunología , Animales , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Proliferación Celular , Células Cultivadas , Células Dendríticas/inmunología , Intolerancia a la Glucosa/inmunología , Inflamación/inmunología , Factor 3 Regulador del Interferón/inmunología , Activación de Linfocitos/inmunología , Antígeno-1 Asociado a Función de Linfocito/biosíntesis , Receptor beta de Linfotoxina/genética , Linfotoxina beta/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/citología , Páncreas/inmunología
3.
Proc Natl Acad Sci U S A ; 109(4): 1210-5, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22167808

RESUMEN

Infections with HIV, hepatitis B virus, and hepatitis C virus can turn into chronic infections, which currently affect more than 500 million patients worldwide. It is generally thought that virus-mediated T-cell exhaustion limits T-cell function, thus promoting chronic disease. Here we demonstrate that natural killer (NK) cells have a negative impact on the development of T-cell immunity by using the murine lymphocytic choriomeningitis virus. NK cell-deficient (Nfil3(-/-), E4BP4(-/-)) mice exhibited a higher virus-specific T-cell response. In addition, NK cell depletion caused enhanced T-cell immunity in WT mice, which led to rapid virus control and prevented chronic infection in lymphocytic choriomeningitis virus clone 13- and reduced viral load in DOCILE-infected animals. Further experiments showed that NKG2D triggered regulatory NK cell functions, which were mediated by perforin, and limited T-cell responses. Therefore, we identified an important role of regulatory NK cells in limiting T-cell immunity during virus infection.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Enfermedad Crónica , Enfermedades Transmisibles/inmunología , Inmunidad Celular/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Virus de la Coriomeningitis Linfocítica , Análisis de Varianza , Animales , Pruebas Inmunológicas de Citotoxicidad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnicas Histológicas , Interferón-alfa/metabolismo , Ratones , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
J Virol ; 83(5): 2226-36, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19073715

RESUMEN

In an earlier report, we provided evidence that expression of CCR5 by primary human T cells renders them permissive for vaccinia virus (VACV) replication. This may represent a mechanism for dissemination throughout the lymphatic system. To test this hypothesis, wild-type CCR5(+/+) and CCR5 null mice were challenged with VACV by intranasal inoculation. In time course studies using different infective doses of VACV, we identified viral replication in the lungs of both CCR5(+/+) and CCR5(-/-) mice, yet there were diminished viral loads in the spleens and brains of CCR5(-/-) mice compared with CCR5(+/+) mice. Moreover, in association with VACV infection, we provide evidence for CD4+ and CD8+ T-cell as well as CD11c+ and F4/80+ cell infiltration into the lungs of CCR5(+/+) but not CCR5(-/-) mice, and we show that the CCR5-expressing T cells harbor virus. We demonstrate that this CCR5 dependence is VACV specific, since CCR5(-/-) mice are as susceptible to intranasal influenza virus (A/WSN/33) infection as CCR5(+/+) mice. In a final series of experiments, we provide evidence that adoptive transfer of CCR5(+/+) bone marrow leukocytes into CCR5(-/-) mice restores VACV permissiveness, with evidence of lung and spleen infection. Taken together, our data suggest a novel role for CCR5 in VACV dissemination in vivo.


Asunto(s)
Pulmón/virología , Receptores CCR5/inmunología , Virus Vaccinia/inmunología , Vaccinia/inmunología , Replicación Viral , Traslado Adoptivo , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/virología , Encéfalo/virología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Femenino , Ratones , Bazo/virología , Vaccinia/virología , Virus Vaccinia/fisiología , Carga Viral
5.
Blood ; 111(10): 4892-901, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18337562

RESUMEN

The multistep, coordinated process of T-cell chemotaxis requires chemokines, and their chemokine receptors, to invoke signaling events to direct cell migration. Here, we examined the role for CCL5-mediated initiation of mRNA translation in CD4(+) T-cell chemotaxis. Using rapamycin, an inhibitor of mTOR, our data show the importance of mTOR in CCL5-mediated T-cell migration. Cycloheximide, but not actinomycin D, significantly reduced chemotaxis, suggesting a possible role for mRNA translation in T-cell migration. CCL5 induced phosphorylation/activation of mTOR, p70 S6K1, and ribosomal protein S6. In addition, CCL5 induced PI-3'K-, phospholipase D (PLD)-, and mTOR-dependent phosphorylation and deactivation of the transcriptional repressor 4E-BP1, which resulted in its dissociation from the eukaryotic initiation factor-4E (eIF4E). Subsequently, eIF4E associated with scaffold protein eIF4G, forming the eIF4F translation initiation complex. Indeed, CCL5 initiated active translation of mRNA, shown by the increased presence of high-molecular-weight polysomes that were significantly reduced by rapamycin treatment. Notably, CCL5 induced protein translation of cyclin D1 and MMP-9, known mediators of migration. Taken together, we describe a novel mechanism by which CCL5 influences translation of rapamycin-sensitive mRNAs and "primes" CD4(+) T cells for efficient chemotaxis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quimiocina CCL5/fisiología , Quimiotaxis de Leucocito , Fosfoproteínas/metabolismo , Biosíntesis de Proteínas , Linfocitos T/fisiología , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Ciclina D1/biosíntesis , Humanos , Metaloproteinasa 9 de la Matriz/biosíntesis , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Proteínas , Linfocitos T/citología , Serina-Treonina Quinasas TOR
6.
Biochem Biophys Res Commun ; 387(2): 381-6, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19607806

RESUMEN

The proliferative capacity of cancer cells is regulated by factors intrinsic to cancer cells and by secreted factors in the microenvironment. Here, we investigated the proto-oncogenic potential of the chemokine receptor, CCR5, in MCF-7 breast cancer cell lines. At physiological levels, CCL5, a ligand for CCR5, enhanced MCF-7.CCR5 proliferation. Treatment with the mTOR inhibitor, rapamycin, inhibited this CCL5-inducible proliferation. Because mTOR directly modulates mRNA translation, we investigated whether CCL5 activation of CCR5 leads to increased translation. CCL5 induced the formation of the eIF4F translation initiation complex through an mTOR-dependent process. Indeed, CCL5 initiated mRNA translation, shown by an increase in high-molecular-weight polysomes. Specifically, we show that CCL5 mediated a rapid up-regulation of protein expression for cyclin D1, c-Myc and Dad-1, without affecting their mRNA levels. Taken together, we describe a mechanism by which CCL5 influences translation of rapamycin-sensitive mRNAs, thereby providing CCR5-positive breast cancer cells with a proliferative advantage.


Asunto(s)
Neoplasias de la Mama/patología , Quimiocina CCL5/metabolismo , Biosíntesis de Proteínas , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Proteínas Reguladoras de la Apoptosis/biosíntesis , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Quimiocina CCL5/genética , Ciclina D1/biosíntesis , Factor 4F Eucariótico de Iniciación/metabolismo , Femenino , Humanos , Proteínas de la Membrana/biosíntesis , Polirribosomas/metabolismo , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Receptores CCR5/metabolismo , Serina-Treonina Quinasas TOR
7.
J Interferon Cytokine Res ; 27(7): 559-65, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17651017

RESUMEN

Signal transducer and activator of transcription 2 (Stat2) is a critical signaling protein involved in mediating interferon-alpha/beta (IFN-alpha/beta) responses. Using site-directed mutagenesis in conjunction with gene microarray and biologic studies, we have previously demonstrated that in addition to Stat2 functioning as a transactivator of transcription of a subset of IFN-inducible genes (ISG), Stat2-DNA binding mediates the transcriptional activation of other ISGs required for IFN-inducible antiviral and growth inhibitory responses. Among these, two candidate genes identified were Jun-D (JUND) and claudin-4 (CLDN4). To further explore the role of JUND and CLDN4 in IFN responses, we conducted knockdown studies using siRNA specific for either JUND or CLDN4 in 2fTGH fibroblast cells, and consistent with cells expressing the DNA-binding mutant of Stat2 (U6A-2VV-II), siRNA-mediated knockdown resulted in cells that exhibited reduced antiproliferative and antiviral responses to IFN. Our data suggest that JUND and CLDN4 are critical mediators of the antiproliferative and antiviral effects of type I IFNs and further confirm the functional importance of the DNA-binding domain of Stat2.


Asunto(s)
ADN/metabolismo , Genes jun , Interferones/metabolismo , Proteínas de la Membrana/genética , Factor de Transcripción STAT2/metabolismo , Línea Celular , Claudina-4 , Regulación de la Expresión Génica , Humanos , Interferones/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor de Transcripción STAT2/genética , Transcripción Genética
8.
Cancer Discov ; 7(11): 1320-1335, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28790030

RESUMEN

Notch activation, which is associated with basal-like breast cancer (BLBC), normally directs tissue patterning, suggesting that it may shape the tumor microenvironment. Here, we show that Notch in tumor cells regulates the expression of two powerful proinflammatory cytokines, IL1ß and CCL2, and the recruitment of tumor-associated macrophages (TAM). Notch also regulates TGFß-mediated activation of tumor cells by TAMs, closing a Notch-dependent paracrine signaling loop between these two cell types. We use a mouse model in which Notch can be regulated in spontaneous mammary carcinoma to confirm that IL1ß and CCL2 production, and macrophage recruitment are Notch-dependent. In human disease, expression array analyses demonstrate a striking association between Notch activation, IL1ß and CCL2 production, macrophage infiltration, and BLBC. These findings place Notch at the nexus of a vicious cycle of macrophage infiltration and amplified cytokine secretion and provide immunotherapeutic opportunities in BLBC.Significance: BLBC is aggressive and has an unmet need for effective targeted treatment. Our data highlight immunotherapeutic opportunities in Notch-activated BLBC. Effective IL1ß and CCL2 antagonists are currently in clinical review to treat benign inflammatory disease, and their transition to the cancer clinic could have a rapid impact. Cancer Discov; 7(11); 1320-35. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1201.


Asunto(s)
Neoplasias de la Mama/genética , Quimiocina CCL2/genética , Interleucina-1beta/genética , Receptor Notch1/genética , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Quimiocina CCL2/inmunología , Quimiocina CCL2/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-1beta/inmunología , Interleucina-1beta/uso terapéutico , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Transgénicos , Comunicación Paracrina/genética , Receptor Notch1/inmunología , Receptor Notch1/uso terapéutico , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/genética , Microambiente Tumoral/genética
9.
Mech Dev ; 122(6): 805-19, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15908185

RESUMEN

Homozygous mice overexpressing Claudin-6 (Cldn6) exhibit a perturbation in the epidermal differentiation program leading to a defective epidermal permeability barrier (EPB) and dehydration induced death ensuing within 48 h of birth [Turksen, K., Troy, T.C., 2002. Permeability barrier dysfunction in transgenic mice overexpressing claudin 6. Development 129, 1775-1784]. Their heterozygous counterparts are also born with an incomplete EPB; however, barrier formation continues after birth and normal hydration levels are achieved by postnatal day 12 allowing survival into adulthood. Heterozygous Inv-Cldn6 mice exhibit a distinct coat phenotype and histological analysis shows mild epidermal hyperkeratosis. Expression of K5 and K14 is aberrant, extending beyond the basal layer into the suprabasal layer where they are not co-localized suggesting that their expression is uncoupled. There is also atypical K17 and patchy K15 expression in the basal layer with no K6 expression in the interfollicular epidermis; together with marked changes in late differentiation markers (e.g. profilaggrin/filaggrin, loricrin, transglutaminase 3) indicating that the normal epidermal differentiation program is modified. The expression compartment of various Cldns is also perturbed although overall protein levels remained comparable. Most notably induction of Cldn5 and Cldn8 was observed in the Inv-Cldn6 epidermis. Heterozygous Inv-Cldn6 animals also exhibit subtle alterations in the differentiation program of the hair follicle including a shorter anagen phase, and altered hair type distribution and length compared to the wild type; the approximately 20% increase in zig-zag hair fibers at the expense of guard hairs and the approximately 30% shorter guard hairs contribute to coat abnormalities in the heterozygous mice. In addition, the transgenic hair follicles exhibit a decreased expression of K15 as well as some hair-specific keratins and express Cldn5 and Cldn18, which are not detectable in the wild type. These data indicate that Cldn6 plays a role in the differentiation processes of the epidermis and hair follicle and supports the notion of a link between Cldn regulation and EPB assembly/maintenance as well as the hair cycle.


Asunto(s)
Epidermis/metabolismo , Regulación del Desarrollo de la Expresión Génica , Folículo Piloso/metabolismo , Proteínas de la Membrana/fisiología , Animales , Western Blotting , Diferenciación Celular , Claudina-5 , Claudinas , Heterocigoto , Homocigoto , Inmunohistoquímica , Queratina-14 , Queratina-15 , Queratina-5 , Queratinas/biosíntesis , Queratinas/metabolismo , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Transgénicos , Fenotipo , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/patología , Factores de Tiempo
10.
Oncoimmunology ; 5(1): e1050575, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26942059

RESUMEN

B7-H4 (B7x, B7S1, VTCN1, DD-0110) is a member of the B7 superfamily that negatively regulates T cell responses.1 In addition, B7-H4 expression is increased on tumors and has been shown to be a negative prognostic marker for many cancers.2 Unexpectedly our recent study demonstrated that B7-H4 inhibited tumor growth and was required to promote effective antitumor responses.3.

11.
Open Biol ; 6(6)2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27335323

RESUMEN

In earlier studies, we showed that CCL5 enhances proliferation and survival of MCF-7 breast cancer cells in an mTOR-dependent manner and we provided evidence that, for T cells, CCL5 activation of CCR5 results in increased glycolysis and enhanced ATP production. Increases in metabolic activity of cancer cells, specifically increased glycolytic activity and increased expression of glucose transporters, are associated with tumour progression. In this report, we provide evidence that CCL5 enhances the proliferation of human breast cancer cell lines (MDA-MB-231, MCF-7) and mouse mammary tumour cells (MMTV-PyMT), mediated by CCR5 activation. Concomitant with enhanced proliferation we show that CCL5 increases cell surface expression of the glucose transporter GLUT1, and increases glucose uptake and ATP production by these cells. Blocking CCL5-inducible glucose uptake abrogates the enhanced proliferation induced by CCL5. We provide evidence that increased glucose uptake is associated with enhanced glycolysis, as measured by extracellular acidification. Moreover, CCL5 enhances the invasive capacity of these breast cancer cells. Using metabolomics, we demonstrate that the metabolic signature of CCL5-treated primary mouse mammary tumour cells reflects increased anabolic metabolism. The implications are that CCL5-CCR5 interactions in the tumour microenvironment regulate metabolic events, specifically glycolysis, to promote tumour proliferation and invasion.


Asunto(s)
Neoplasias de la Mama/metabolismo , Quimiocina CCL5/metabolismo , Glucólisis , Metabolómica/métodos , Receptores CCR5/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Células MCF-7 , Ratones
12.
Methods Mol Biol ; 289: 113-20, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15502176

RESUMEN

Immunohistochemistry is an efficient means of localizing specific proteins to their relative expression compartment in the epidermis thereby providing evidence as to their functionality. This chapter therefore describes a dependable method for immunolocalization within the epidermis.


Asunto(s)
Células Epidérmicas , Queratinocitos/citología , Queratinas/metabolismo , Piel/citología , Animales , Epidermis/metabolismo , Inmunohistoquímica , Queratinocitos/metabolismo , Ratones , Piel/metabolismo
13.
Cancer Immunol Res ; 3(2): 184-95, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25527357

RESUMEN

The B7 family plays a critical role in both positive and negative regulation of immune responses by engaging a variety of receptors on lymphocytes. Importantly, blocking coinhibitory molecules using antibodies specific for CTLA-4 and PD-1 enhances tumor immunity in a subset of patients. Therefore, it is critical to understand the role of different B7 family members since they may be suitable therapeutic targets. B7-H4 is another member that inhibits T-cell function, and it is also upregulated on a variety of tumors and has been proposed to promote tumor growth. Here, we investigate the role of B7-H4 in tumor development and show that B7-H4 expression inhibits tumor growth in two mouse models. Furthermore, we show that B7-H4 expression is required for antitumor immune responses in a mouse model of mammary tumorigenesis. We found that the expression levels of B7-H4 correlate with MHC class I expression in both mouse and human samples. We show that IFNγ upregulates B7-H4 expression on mouse embryo fibroblasts and that the upregulation of B7-H4 on tumors is dependent on T cells. Notably, patients with breast cancer with increased B7-H4 expression show a prolonged time to recurrence. These studies demonstrate a positive role for B7-H4 in promoting antitumor immunity.


Asunto(s)
Neoplasias Mamarias Experimentales/inmunología , Microambiente Tumoral/inmunología , Inhibidor 1 de la Activación de Células T con Dominio V-Set/inmunología , Animales , Biomarcadores de Tumor/metabolismo , Citotoxicidad Inmunológica/inmunología , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Granzimas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunidad Celular , Interferón gamma/biosíntesis , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/prevención & control , Ratones Transgénicos , Proteínas de Neoplasias/inmunología , Linfocitos T Citotóxicos/enzimología , Linfocitos T Citotóxicos/inmunología , Regulación hacia Arriba/inmunología , Inhibidor 1 de la Activación de Células T con Dominio V-Set/deficiencia , Inhibidor 1 de la Activación de Células T con Dominio V-Set/genética
14.
J Interferon Cytokine Res ; 32(3): 127-37, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22280104

RESUMEN

In earlier studies we provided evidence that vaccinia virus (VACV) phosphorylation-activation of host cell signaling effectors is critical for subsequent viral replication. In this report, using mass spectrometry-based proteomics, we have identified 387 host cell proteins that co-immunoprecipitate with VACV in infected, permissive PM1.CCR5 human T cells. Among these, glomulin was distinguishable based on its known interaction with a tyrosine kinase receptor, c-Met, its ability to become tyrosine-phosphorylated, and its association with signaling effectors. siRNA knockdown of glomulin expression in PM1.CCR5 T cells reduces VACV infection. Glomulin interacts with the inactive, nonphosphorylated form of c-MET. We demonstrate that treatment of PM1.CCR5 T cells with a c-Met phosphorylation inhibitor leads to a significant reduction in VACV infectivity. Additionally, inhibition of phosphorylation of c-Met abrogates VACV-inducible phosphorylation of Erk 1/2 and IRS-2, signaling effectors identified as critical for VACV infection. These data identify glomulin as a permissivity factor for VACV infection and as a potential therapeutic target for inhibition of VACV infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Virus Vaccinia/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular , Silenciador del Gen , Humanos , Indoles/farmacología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Proteómica/métodos , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Sulfonamidas/farmacología , Tirosina/metabolismo
15.
J Interferon Cytokine Res ; 31(7): 545-52, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21323567

RESUMEN

Type I interferons (IFNs) exhibit antiproliferative activity and apoptotic effects, and regulate an immune response by activating multiple cells types, including dendritic cells, cytotoxic T cells, and natural killer cells. Most recently, a report in the literature identified dysfunctional induction of a type I IFN response in cancer stem cells--specifically, breast cancer-initiating cells, implicating this defect in progression to breast cancer. Indeed, accumulating evidence suggests that cancer stem cells/cancer-initiating cells are prevalent in leukemias and solid tumors, are resistant to chemotherapy and radiation therapy, and therefore likely contribute to tumor recurrence. IFN-ß treatment of human glioma xenografts leads to disruption of the vascular niche of glioma stem cells, in further support of a potential therapeutic effect of IFN treatment in limiting cancer stem cells. The implications are that restoring an IFN response, or enhancing an IFN response, may invoke a reduction, or elimination of both cancer stem cells and tumor cells. In this review, the clinical application of type I IFNs, mainly IFN-αs, will be reviewed.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Interferón Tipo I/metabolismo , Leucemia/tratamiento farmacológico , Células Madre Neoplásicas/metabolismo , Animales , Neoplasias de la Mama/inmunología , Resistencia a Medicamentos/genética , Quimioterapia/tendencias , Femenino , Predicción , Humanos , Interferón Tipo I/genética , Leucemia/inmunología , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Nicho de Células Madre/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Antiviral Res ; 89(1): 75-82, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21093489

RESUMEN

Interferons (IFNs) are a first line of defense against viral infection. Herein we describe the use of an adenovirus vectored mouse IFN alpha gene (mDEF201) as a prophylactic and treatment countermeasure in a SARS-CoV-infected BALB/c mouse model. Complete survival protection was observed in mice given a single dose of mDEF201 administered intranasally 1, 3, 5, 7, or 14 days prior to lethal SARS-CoV challenge (p<0.001), and body weights of these treated mice were unaffected by the challenge. In addition, low doses of mDEF201 protected lungs in a dose dependent manner as measured by a reduction in gross pathology. Intranasal treatment with mDEF201 ranging from 10(6) to 10(8)PFU significantly protected mice against a lethal SARS-CoV infection in a dose dependent manner up to 12h post infection (p<0.001). The data suggest that mDEF201 is a new class of antiviral agent further development as treatment for SARS-CoV infections.


Asunto(s)
Antivirales/administración & dosificación , Productos Biológicos/administración & dosificación , Interferón-alfa/administración & dosificación , Síndrome Respiratorio Agudo Grave/prevención & control , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Adenoviridae/genética , Administración Intranasal , Animales , Antivirales/inmunología , Productos Biológicos/genética , Productos Biológicos/inmunología , Peso Corporal , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos , Humanos , Interferón-alfa/genética , Interferón-alfa/inmunología , Ratones , Ratones Endogámicos BALB C , Síndrome Respiratorio Agudo Grave/mortalidad , Síndrome Respiratorio Agudo Grave/patología , Análisis de Supervivencia , Células Vero
17.
PLoS One ; 5(11): e13927, 2010 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21085662

RESUMEN

Type I interferons (IFNs) function as the first line of defense against viral infections by modulating cell growth, establishing an antiviral state and influencing the activation of various immune cells. Viruses such as influenza have developed mechanisms to evade this defense mechanism and during infection with influenza A viruses, the non-structural protein 1 (NS1) encoded by the virus genome suppresses induction of IFNs-α/ß. Here we show that expression of avian H5N1 NS1 in HeLa cells leads to a block in IFN signaling. H5N1 NS1 reduces IFN-inducible tyrosine phosphorylation of STAT1, STAT2 and STAT3 and inhibits the nuclear translocation of phospho-STAT2 and the formation of IFN-inducible STAT1:1-, STAT1:3- and STAT3:3- DNA complexes. Inhibition of IFN-inducible STAT signaling by NS1 in HeLa cells is, in part, a consequence of NS1-mediated inhibition of expression of the IFN receptor subunit, IFNAR1. In support of this NS1-mediated inhibition, we observed a reduction in expression of ifnar1 in ex vivo human non-tumor lung tissues infected with H5N1 and H1N1 viruses. Moreover, H1N1 and H5N1 virus infection of human monocyte-derived macrophages led to inhibition of both ifnar1 and ifnar2 expression. In addition, NS1 expression induces up-regulation of the JAK/STAT inhibitors, SOCS1 and SOCS3. By contrast, treatment of ex vivo human lung tissues with IFN-α results in the up-regulation of a number of IFN-stimulated genes and inhibits both H5N1 and H1N1 virus replication. The data suggest that NS1 can directly interfere with IFN signaling to enhance viral replication, but that treatment with IFN can nevertheless override these inhibitory effects to block H5N1 and H1N1 virus infections.


Asunto(s)
Virus de la Influenza A/fisiología , Interferones/metabolismo , Transducción de Señal/fisiología , Proteínas no Estructurales Virales/fisiología , Células Cultivadas , Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Subtipo H5N1 del Virus de la Influenza A/fisiología , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Interferones/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/virología , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/virología , Microscopía Confocal , Fosforilación , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Técnicas de Cultivo de Tejidos , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
18.
J Biol Chem ; 281(35): 25184-94, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16807236

RESUMEN

CCL5 (RANTES (regulated on activation normal T cell expressed and secreted)) and its cognate receptor, CCR5, have been implicated in T cell activation. CCL5 binding to glycosaminoglycans (GAGs) on the cell surface or in extracellular matrix sequesters CCL5, thereby immobilizing CCL5 to provide the directional signal. In two CCR5-expressing human T cell lines, PM1.CCR5 and MOLT4.CCR5, and in human peripheral blood-derived T cells, micromolar concentrations of CCL5 induce apoptosis. CCL5-induced cell death involves the cytosolic release of cytochrome c, the activation of caspase-9 and caspase-3, and poly(ADP-ribose) polymerase cleavage. CCL5-induced apoptosis is CCR5-dependent, since native PM1 and MOLT4 cells lacking CCR5 expression are resistant to CCL5-induced cell death. Furthermore, we implicate tyrosine 339 as a critical residue involved in CCL5-induced apoptosis, since PM1 cells expressing a tyrosine mutant receptor, CCR5Y339F, do not undergo apoptosis. We show that CCL5-CCR5-mediated apoptosis is dependent on cell surface GAG binding. The addition of exogenous heparin and chondroitin sulfate and GAG digestion from the cell surface protect cells from apoptosis. Moreover, the non-GAG binding variant, (44AANA47)-CCL5, fails to induce apoptosis. To address the role of aggregation in CCL5-mediated apoptosis, nonaggregating CCL5 mutant E66S, which forms dimers, and E26A, which form tetramers at micromolar concentrations, were utilized. Unlike native CCL5, the E66S mutant fails to induce apoptosis, suggesting that tetramers are the minimal higher ordered CCL5 aggregates required for CCL5-induced apoptosis. Viewed altogether, these data suggest that CCL5-GAG binding and CCL5 aggregation are important for CCL5 activity in T cells, specifically in the context of CCR5-mediated apoptosis.


Asunto(s)
Apoptosis , Quimiocinas CC/fisiología , Glicosaminoglicanos/metabolismo , Receptores CCR5/fisiología , Linfocitos T/metabolismo , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Quimiocina CCL5 , Citocromos c/metabolismo , Matriz Extracelular/metabolismo , Humanos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Unión Proteica , Linfocitos T/patología
19.
J Virol ; 80(14): 7245-59, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16809330

RESUMEN

Vaccinia virus, a poxvirus, produces structurally distinct forms of virions for which the immediate events following cell entry are ill-defined. We provide evidence that intracellular mature virus (IMV) enters both permissive and nonpermissive T-cell lines and that introduction of CCR5 into nonpermissive mouse fibroblasts or human primary T cells renders the cells permissive for vaccinia replication. Notably, T cells expressing CCR5 in which tyrosine 339 in the intracellular region is replaced by phenylalanine no longer support virus replication or virus-inducible activation of specific host cell signaling effectors IRS-2, Grb2, and Erk1/2. We show that following IMV entry into the cell, the intact but not the tyrosine-deficient CCR5 is rapidly internalized and colocalizes with virus. This colocalization precedes virus-inducible signaling and replication.


Asunto(s)
Fibroblastos/metabolismo , Receptores CCR5/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Virus Vaccinia/fisiología , Replicación Viral/fisiología , Sustitución de Aminoácidos , Animales , Fibroblastos/virología , Proteína Adaptadora GRB2/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Células 3T3 NIH , Fosfoproteínas/metabolismo , Fosforilación , Mutación Puntual , Procesamiento Proteico-Postraduccional/genética , Receptores CCR5/genética , Linfocitos T/virología , Tirosina/genética , Tirosina/metabolismo , Vaccinia/genética , Vaccinia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA