Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
Nat Commun ; 9(1): 3000, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30050138

RESUMO

The original version of this Article omitted a declaration from the competing interests statement, which should have included the following: 'R.D.S. is a cofounder, stock holder, and scientific advisory board member of Jounce Therapeutics and Neon Therapeutics, and a member of the scientific advisory boards of BioLegend, Constellation, Lytix, and NGM. He also received research funding from Janssen and Agios.'. This has now been corrected in both the PDF and HTML versions of the Article.

2.
Nat Commun ; 8(1): 562, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28916749

RESUMO

The analysis of neoantigen-specific CD8+ T cells in tumour-bearing individuals is challenging due to the small pool of tumour antigen-specific T cells. Here we show that mass cytometry with multiplex combinatorial tetramer staining can identify and characterize neoantigen-specific CD8+ T cells in mice bearing T3 methylcholanthrene-induced sarcomas that are susceptible to checkpoint blockade immunotherapy. Among 81 candidate antigens tested, we identify T cells restricted to two known neoantigens simultaneously in tumours, spleens and lymph nodes in tumour-bearing mice. High-dimensional phenotypic profiling reveals that antigen-specific, tumour-infiltrating T cells are highly heterogeneous. We further show that neoantigen-specific T cells display a different phenotypic profile in mice treated with anti-CTLA-4 or anti-PD-1 immunotherapy, whereas their peripheral counterparts are not affected by the treatments. Our results provide insights into the nature of neoantigen-specific T cells and the effects of checkpoint blockade immunotherapy.Immune checkpoint blockade (ICB) therapies can unleash anti-tumour T-cell responses. Here the authors show, by integrating MHC tetramer multiplexing, mass cytometry and high-dimensional analyses, that neoantigen-specific, tumour-infiltrating T cells are highly heterogeneous and are subjected to ICB modulations.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Sarcoma Experimental/imunologia , Animais , Antineoplásicos Imunológicos/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Antígeno CTLA-4/antagonistas & inibidores , Imunofenotipagem , Imunoterapia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Metilcolantreno/toxicidade , Camundongos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Sarcoma Experimental/induzido quimicamente
3.
Cell Death Differ ; 21(2): 234-46, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24037089

RESUMO

We previously reported that STAT1 expression is frequently abrogated in human estrogen receptor-α-positive (ERα(+)) breast cancers and mice lacking STAT1 spontaneously develop ERα(+) mammary tumors. However, the precise mechanism by which STAT1 suppresses mammary gland tumorigenesis has not been fully elucidated. Here we show that STAT1-deficient mammary epithelial cells (MECs) display persistent prolactin receptor (PrlR) signaling, resulting in activation of JAK2, STAT3 and STAT5A/5B, expansion of CD61(+) luminal progenitor cells and development of ERα(+) mammary tumors. A failure to upregulate SOCS1, a STAT1-induced inhibitor of JAK2, leads to unopposed oncogenic PrlR signaling in STAT1(-/-) MECs. Prophylactic use of a pharmacological JAK2 inhibitor restrains the proportion of luminal progenitors and prevents disease induction. Systemic inhibition of activated JAK2 induces tumor cell death and produces therapeutic regression of pre-existing endocrine-sensitive and refractory mammary tumors. Thus, STAT1 suppresses tumor formation in mammary glands by preventing the natural developmental function of a growth factor signaling pathway from becoming pro-oncogenic. In addition, targeted inhibition of JAK2 may have significant therapeutic potential in controlling ERα(+) breast cancer in humans.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Janus Quinase 2/metabolismo , Neoplasias Mamárias Animais/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fator de Transcrição STAT1/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Compostos Heterocíclicos com 3 Anéis/farmacologia , Janus Quinase 2/antagonistas & inibidores , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/genética , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Fator de Transcrição STAT1/deficiência , Transdução de Sinais/efeitos dos fármacos , Proteína 1 Supressora da Sinalização de Citocina
4.
EMBO J ; 20(19): 5431-42, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11574475

RESUMO

A tripartite receptor comprising the external region of the erythropoietin (Epo) receptor, the transmembrane and JAK-binding domains of the gp130 subunit of the interleukin-6 (IL-6) receptor, and a seven amino acid STAT1 recruitment motif (Y440) from the interferon (IFN)-gamma receptor, efficiently mediates an IFN-gamma-like response. An analogous completely foreign chimeric receptor in which the Y440 motif is replaced with the Y905 motif from gp130 also mediates an IFN-gamma-like response, but less efficiently. The IFNGR1 signal-transducing subunit of the IFN-gamma receptor is tyrosine phosphorylated through the chimeric receptors and the endogenous IL-6 and OSM receptors. Cross phosphorylation of IFNGR1 is not, however, required for the IFN-gamma-like response through the chimeric receptors, nor does it mediate an IFN-gamma-like response to IL-6 or OSM. The data argue strongly for modular JAK/STAT signalling and against any rigid structural organization for the "pathways" involved. They emphasize the likely high degree of overlap between the signals generated from disparate JAK-receptor complexes and show that relatively minor changes in such complexes can profoundly affect the response.


Assuntos
Interferon gama/metabolismo , Proteínas Nucleares , Receptores Imunológicos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/biossíntese , Humanos , Receptor de Interferon alfa e beta , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Receptores Imunológicos/genética , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Fator de Transcrição STAT1 , Transdução de Sinais , Transativadores/biossíntese , Transativadores/metabolismo , Receptor de Interferon gama
5.
Cancer Res ; 61(17): 6540-7, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11522652

RESUMO

IRF9/p48/ISGF3gamma (IRF9) is an IFN regulatory factor that mediates signaling by type I IFNs (IFNalpha and IFNbeta). After single-step selection of breast adenocarcinoma cells in paclitaxel, differential display and single gene analysis demonstrated that transcriptional activation of IRF9 and other IFN-responsive genes, independent of IFN, corresponded with resistance to antimicrotubule agents. Transient overexpression of IRF9 reproduced the drug-resistance phenotype and induced expression of IFN-responsive genes. However, drug resistance was not induced by overexpression of Stat1 or Stat2, or treatment with IFNalpha per se. Using a donor-matched array of cDNA prepared from human tumor and normal tissue from a variety of organs, we observed overexpression of IRF9 in approximately one-half of breast and uterine tumors, which indicated that IRF9 may be important in signaling in these tumor types. These data identify a novel IFN-independent role for IRF9 in the development of resistance to antimicrotubule agents in breast tumor cells and may link downstream mediators of IFN signaling to drug resistance in human cancers.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Proteínas de Ligação a DNA/fisiologia , Paclitaxel/farmacologia , Fatores de Transcrição/fisiologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Ciclo Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Interferon Tipo I/fisiologia , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Microtúbulos/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Vimblastina/farmacologia
6.
Science ; 293(5528): 300-3, 2001 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-11452125

RESUMO

Interferons (IFN) alpha/beta and gamma induce the formation of two transcriptional activators: gamma-activating factor (GAF) and interferon-stimulated gamma factor 3 (ISGF3). We report a natural heterozygous germline STAT1 mutation associated with susceptibility to mycobacterial but not viral disease. This mutation causes a loss of GAF and ISGF3 activation but is dominant for one cellular phenotype and recessive for the other. It impairs the nuclear accumulation of GAF but not of ISGF3 in heterozygous cells stimulated by IFNs. Thus, the antimycobacterial, but not the antiviral, effects of human IFNs are principally mediated by GAF.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Mutação em Linhagem Germinativa , Imunidade , Interferon-alfa/imunologia , Interferon gama/imunologia , Infecções por Mycobacterium/imunologia , Transativadores/fisiologia , Viroses/imunologia , Adulto , Animais , Linhagem Celular , Criança , Pré-Escolar , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação da Expressão Gênica , Humanos , Imunidade/genética , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Interferon-alfa/metabolismo , Interferon gama/metabolismo , Janus Quinase 1 , Camundongos , Infecções por Mycobacterium/genética , Complexo Mycobacterium avium/imunologia , Infecção por Mycobacterium avium-intracellulare/genética , Infecção por Mycobacterium avium-intracellulare/imunologia , Mycobacterium bovis , Linhagem , Ligação Proteica , Proteínas Tirosina Quinases/genética , Fator de Transcrição STAT1 , Transdução de Sinais , Vírus 40 dos Símios , Transativadores/genética , Fatores de Transcrição/fisiologia , Viroses/genética
7.
Proc Natl Acad Sci U S A ; 98(12): 6674-9, 2001 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-11390994

RESUMO

Although Stat1 is essential for cells to respond fully to IFN-gamma, there is substantial evidence that, in the absence of Stat1, IFN-gamma can still regulate the expression of some genes, induce an antiviral state and affect cell growth. We have now identified many genes that are regulated by IFN-gamma in serum-starved Stat1-null mouse fibroblasts. The proteins induced by IFN-gamma in Stat1-null cells can account for the substantial biological responses that remain. Some genes are induced in both wild-type and Stat1-null cells and thus are truly Stat1-independent. Others are subject to more complex regulation in response to IFN-gamma, repressed by Stat1 in wild-type cells and activated in Stat1-null cells. Many genes induced by IFN-gamma in Stat1-null fibroblasts also are induced by platelet-derived growth factor in wild-type cells and thus are likely to be involved in cell proliferation. In mouse cells expressing the docking site mutant Y440F of human IFN-gamma receptor subunit 1, the mouse Stat1 is not phosphorylated in response to human IFN-gamma, but c-myc and c-jun are still induced, showing that the Stat1 docking site is not required for Stat1-independent signaling.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Imediatamente Precoces , Interferon gama/farmacologia , Transativadores/fisiologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/biossíntese , Células Cultivadas , Quimiocinas/genética , Proteínas de Ligação a DNA/biossíntese , Proteína 1 de Resposta de Crescimento Precoce , Genes Precoces , Genes jun , Genes myc , Humanos , Camundongos , Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores de Interferon/fisiologia , Fator de Transcrição STAT1 , Fatores de Transcrição/biossíntese , Receptor de Interferon gama
8.
Proc Natl Acad Sci U S A ; 98(12): 6680-5, 2001 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-11390995

RESUMO

Although Stat1 is required for many IFN-dependent responses, recent work has shown that IFNgamma functions independently of Stat1 to affect the growth of tumor cells or immortalized fibroblasts. We now demonstrate that both IFNgamma and IFNalpha/beta regulate proliferative responses in cells of the mononuclear phagocyte lineage derived from Stat1-null mice. Using both representational difference analysis and gene arrays, we show that IFNgamma exerts its Stat1-independent actions on mononuclear phagocytes by regulating the expression of many genes. This result was confirmed by monitoring changes in expression and function of the corresponding gene products. Regulation of the expression of these genes requires the IFNgamma receptor and Jak1. The physiologic relevance of IFN-dependent, Stat1-independent signaling was demonstrated by monitoring antiviral responses in Stat1-null mice. Thus, the IFN receptors engage alternative Stat1-independent signaling pathways that have important physiological consequences.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Interferons/farmacologia , Transativadores/fisiologia , Animais , Divisão Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Janus Quinase 1 , Macrófagos/metabolismo , Camundongos , Proteínas Tirosina Quinases/fisiologia , Receptores de Interferon/fisiologia , Fator de Transcrição STAT1 , Receptor de Interferon gama
9.
Nature ; 410(6832): 1107-11, 2001 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-11323675

RESUMO

Lymphocytes were originally thought to form the basis of a 'cancer immunosurveillance' process that protects immunocompetent hosts against primary tumour development, but this idea was largely abandoned when no differences in primary tumour development were found between athymic nude mice and syngeneic wild-type mice. However, subsequent observations that nude mice do not completely lack functional T cells and that two components of the immune system-IFNgamma and perforin-help to prevent tumour formation in mice have led to renewed interest in a tumour-suppressor role for the immune response. Here we show that lymphocytes and IFNgamma collaborate to protect against development of carcinogen-induced sarcomas and spontaneous epithelial carcinomas and also to select for tumour cells with reduced immunogenicity. The immune response thus functions as an effective extrinsic tumour-suppressor system. However, this process also leads to the immunoselection of tumour cells that are more capable of surviving in an immunocompetent host, which explains the apparent paradox of tumour formation in immunologically intact individuals.


Assuntos
Carcinoma/imunologia , Interferon gama/imunologia , Linfócitos/imunologia , Sarcoma Experimental/imunologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/imunologia , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Feminino , Antígenos H-2/imunologia , Imunocompetência , Imunofenotipagem , Metilcolantreno , Camundongos , Transplante de Neoplasias , Retroviridae/isolamento & purificação
10.
Science ; 291(5511): 2162-5, 2001 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-11251123

RESUMO

The role of NF-kappaB-inducing kinase (NIK) in cytokine signaling remains controversial. To identify the physiologic functions of NIK, we disrupted the NIK locus by gene targeting. Although NIK-/- mice displayed abnormalities in both lymphoid tissue development and antibody responses, NIK-/- cells manifested normal NF-kappaB DNA binding activity when treated with a variety of cytokines, including tumor necrosis factor (TNF), interleukin-1 (IL-1), and lymphotoxin-beta (LTbeta). However, NIK was selectively required for gene transcription induced through ligation of LTbeta receptor but not TNF receptors. These results reveal that NIK regulates the transcriptional activity of NF-kappaB in a receptor-restricted manner.


Assuntos
NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Transcrição Gênica , Animais , Anticorpos Monoclonais , Linfócitos B/metabolismo , Células Cultivadas , DNA/metabolismo , Fibroblastos/metabolismo , Marcação de Genes , Genes Reporter , Interleucina-1/metabolismo , Interleucina-1/farmacologia , Ligantes , Tecido Linfoide/anormalidades , Receptor beta de Linfotoxina , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , Proteínas Serina-Treonina Quinases/genética , Receptores do Fator de Necrose Tumoral/imunologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Quinase Induzida por NF-kappaB
11.
J Biol Chem ; 276(1): 287-97, 2001 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-10995751

RESUMO

Interferons (IFNs) regulate the expression of a number of cellular genes by activating the JAK-STAT pathway. We have recently discovered that CCAAAT/enhancer-binding protein-beta (C/EBP-beta) induces gene transcription through a novel IFN response element called the gamma-IFN-activated transcriptional element (Roy, S. K., Wachira, S. J., Weihua, X., Hu, J., and Kalvakolanu, D. V. (2000) J. Biol. Chem. 275, 12626-12632. Here, we describe a new IFN-gamma-stimulated pathway that operates C/EBP-beta-regulated gene expression independent of JAK1. We show that ERKs are activated by IFN-gamma to stimulate C/EBP-beta-dependent expression. Sustained ERK activation directly correlated with C/EBP-beta-dependent gene expression in response to IFN-gamma. Mutant MKK1, its inhibitors, and mutant ERK suppressed IFN-gamma-stimulated gene induction through the gamma-IFN-activated transcriptional element. Ras and Raf activation was not required for this process. Furthermore, Raf-1 phosphorylation negatively correlated with its activity. Interestingly, C/EBP-beta-induced gene expression required STAT1, but not JAK1. A C/EBP-beta mutant lacking the ERK phosphorylation site failed to promote IFN-stimulated gene expression. Thus, our data link C/EBP-beta to IFN-gamma signaling through ERKs.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon gama/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Linhagem Celular , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática/efeitos dos fármacos , Genes Reporter , Interferon gama/antagonistas & inibidores , Interferon gama/genética , Janus Quinase 1 , MAP Quinase Quinase 1 , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/genética , Mutação , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-raf/fisiologia , Proteínas Recombinantes , Fator de Transcrição STAT1 , Transativadores/deficiência , Transativadores/genética , Transativadores/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno
12.
Curr Protoc Immunol ; Chapter 6: Unit 6.8, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18432821

RESUMO

This unit describes two protocols that can be used to quantitate interferon gamma (IFN-gamma), an important modulatory lymphokine that regulates natural, cell-mediated, and humoral immunity by eliciting a number of biological responses in many different cell types. Depending upon the specificity of the reagents used, the assays will detect either human or murine IFN-gamma. The Basic Protocol is an enzyme-linked immunosorbent assay (ELISA) based on IFN-gamma's unique antigenic structure. The Alternate Protocol is a functional activity assay based on IFN-gamma's ability to induce major histocompatibility (MHC) class II antigens (also termed Ia antigens) on responsive cells. The advantage of the ELISA protocol is its specificity, while the advantage of the MHC protocol is its sensitivity.


Assuntos
Bioensaio/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Antígenos de Histocompatibilidade Classe II/biossíntese , Interferon gama/análise , Animais , Linhagem Celular , Humanos , Camundongos
13.
Immunity ; 13(2): 265-76, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10981969

RESUMO

We generated the DUC18 T cell receptor transgenic mouse expressing an H-2Kd -restricted transgenic T cell receptor specific for the syngeneic CMS5 fibrosarcoma rejection antigen mutated ERK2(136-144). DUC18 mice were capable of specifically eliminating lethal CMS5 tumor challenges, and transfer of DUC18 splenocytes to naive nontransgenic recipients conferred protection from subsequent and established CMS5 tumor burdens. Eradication of established tumor burdens by adoptive transfer of DUC18 splenocytes was dose and time dependent. Transferred tumor-specific T cells remained functional in vivo and capable of rejecting small tumors even in the presence of large, established tumor burdens. These findings highlight the kinetic battle between tumor growth and the production of a tumor-specific response and have critical implications for effective adoptive immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica , Fibrossarcoma/imunologia , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/transplante , Fibrossarcoma/terapia , Camundongos , Camundongos Transgênicos , Mutação , Receptores de Antígenos de Linfócitos T/genética
14.
J Immunol ; 165(5): 2362-6, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10946258

RESUMO

Type I IFNs, IFN-alpha, -beta, and -omega, are cytokine family members with multiple immune response roles, including the promotion of cell growth and differentiation. Conversely, the type I IFNs are potent inhibitors of IL-7-dependent growth of early B lineage progenitors, effectively aborting further B lineage differentiation at the pro-B cell stage. Type I IFNs alpha and beta function via receptor-mediated activation of a Jak/Stat signaling pathway in which Stat-1 is functionally important, because many IFN-induced responses are abrogated in Stat-1-deficient mice. To the contrary, we show here that the inhibition of IL-7-dependent B lymphopoiesis by IFN-alphabeta is unaffected in Stat-1-deficient mice. The present data indicate that the type I IFNs can activate an alternative signaling pathway in which neither Stat-1 nor phosphatidylinositol 3'-kinase are essential components.


Assuntos
Linfócitos B/citologia , Linfócitos B/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Inibidores do Crescimento/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Interferon Tipo I/fisiologia , Transativadores/deficiência , Transativadores/genética , Animais , Antígenos Ly/biossíntese , Linfócitos B/enzimologia , Linfócitos B/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Divisão Celular/genética , Divisão Celular/imunologia , Linhagem Celular , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Proteínas de Ligação a DNA/fisiologia , Feminino , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Hematopoéticas/imunologia , Interleucina-7/fisiologia , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fosfatidilinositol 3-Quinases/fisiologia , Fator de Transcrição STAT1 , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Transativadores/fisiologia , Regulação para Cima/genética , Regulação para Cima/imunologia
15.
J Infect Dis ; 181(1): 379-84, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10608793

RESUMO

Complete deficiency of either of the two human interferon (IFN)-gamma receptor components, the ligand-binding IFN-gammaR1 chain and the signaling IFN-gammaR2 chain, is invariably associated with early-onset infection caused by bacille Calmette-Guérin vaccines and/or environmental nontuberculous mycobacteria, poor granuloma formation, and a fatal outcome in childhood. Partial IFN-gammaR1 deficiency is associated with a milder histopathologic and clinical phenotype. Cells from a 20-year-old healthy person with a history of curable infections due to bacille Calmette-Guérin and Mycobacterium abscessus and mature granulomas in childhood were investigated. There was a homozygous nucleotide substitution in IFNGR2, causing an amino acid substitution in the extracellular region of the encoded receptor. Cell surface IFN-gammaR2 were detected by flow cytometry. Cellular responses to IFN-gamma were impaired but not abolished. Transfection with the wild-type IFNGR2 gene restored full responsiveness to IFN-gamma. This is the first demonstration of partial IFN-gammaR2 deficiency in humans.


Assuntos
Vacina BCG/efeitos adversos , Infecções por Mycobacterium/imunologia , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Adulto , Proteínas de Ligação a DNA/metabolismo , Feminino , Genótipo , Antígenos HLA-DR/metabolismo , Homozigoto , Humanos , Mycobacterium bovis/imunologia , Mycobacterium bovis/patogenicidade , Linhagem , Fenótipo , Mutação Puntual , Fator de Transcrição STAT1 , Transdução de Sinais , Transativadores/metabolismo , Receptor de Interferon gama
16.
Nat Med ; 5(12): 1370-4, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10581078

RESUMO

At present, little is known about the pathogenesis of acute virus-induced shock and pulmonary failure. A chief impediment in understanding the underlying disease mechanisms and developing treatment strategies has been the lack of a suitable animal model. This study describes a mouse model of virus-induced systemic shock and respiratory distress, and shows that blockade of the lymphotoxin beta receptor pathway reverses the disease.


Assuntos
Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Insuficiência Respiratória/terapia , Choque Séptico/terapia , Animais , Anticorpos Monoclonais/farmacologia , Modelos Animais de Doenças , Feminino , Humanos , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Coriomeningite Linfocítica/terapia , Receptor beta de Linfotoxina , Masculino , Camundongos , Camundongos Endogâmicos NZB , Insuficiência Respiratória/imunologia , Insuficiência Respiratória/patologia , Choque Séptico/imunologia , Choque Séptico/patologia , Transdução de Sinais , Fatores de Tempo
17.
J Virol ; 73(12): 9944-51, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10559307

RESUMO

Herein, we report that Autographa californica nucleopolyhedrovirus, a member of the Baculoviridae family, is capable of stimulating antiviral activity in mammalian cells. Baculoviruses are not pathogenic to mammalian cells. Nevertheless, live baculovirus is shown here to induce interferons (IFN) from murine and human cell lines and induces in vivo protection of mice from encephalomyocarditis virus infection. Monoclonal antibodies specific for the baculovirus envelope gp67 neutralize baculovirus-dependent IFN production. Moreover, UV treatment of baculovirus eliminates both infectivity and IFN-inducing activity. In contrast, the IFN-inducing activity of the baculovirus was unaffected by DNase or RNase treatment. These data demonstrate that IFN production can be induced in mammalian cells by baculovirus even though the cells fail to serve as a natural host for an active viral infection. Baculoviruses, therefore, provide a novel model in which to study at least one alternative mechanism for IFN induction in mammalian cells.


Assuntos
Interferon-alfa/imunologia , Interferon beta/imunologia , Nucleopoliedrovírus/imunologia , Animais , Anticorpos Monoclonais/imunologia , Células COS , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/prevenção & controle , DNA/imunologia , Feminino , Humanos , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Lipopolissacarídeos/imunologia , Mamíferos , Vírus Elberfeld do Camundongo/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Mariposas/virologia , Testes de Neutralização , RNA de Cadeia Dupla/imunologia
18.
Proc Natl Acad Sci U S A ; 96(15): 8633-8, 1999 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-10411927

RESUMO

CD4(+) T cells can eliminate tumor cells in vivo in the absence of CD8(+) T cells. We have CD4(+) T cells specific for a MHC class II-restricted, tumor-specific peptide derived from a mutant ribosomal protein expressed by the UV light-induced tumor 6132A-PRO. By using neutralizing mAb specific for murine IFN-gamma and adoptive transfer of CD4(+) T cells into severe combined immunodeficient mice, we show that anti-IFN-gamma treatment abolishes the CD4(+) T cell-mediated rejection of the tumor cells in vivo. The tumor cells were MHC class II negative, and IFN-gamma did not induce MHC class II expression in vitro. Therefore, the tumor-specific antigenic peptide must be presented by host cells and not the tumor cells. Tumor cells transduced to secrete IFN-gamma had a markedly reduced growth rate in severe combined immunodeficient mice, but IFN-gamma did not inhibit the growth of the tumor cells in vitro. Furthermore, tumor cells stably expressing a dominant-negative truncated form of the murine IFN-gamma receptor alpha chain, and therefore insensitive to IFN-gamma, nevertheless were rejected by the adoptively transferred CD4(+) T cells. Thus, host cells, and not tumor cells, seem to be the target of IFN-gamma. Together, these results show that CD4(+) T cells can eliminate IFN-gamma-insensitive, MHC class II-negative cancer cells by an indirect mechanism that depends on IFN-gamma.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/análise , Interferon gama/metabolismo , Células Tumorais Cultivadas/imunologia , Sequência de Aminoácidos , Animais , Divisão Celular , Células Cultivadas , Citometria de Fluxo , Genes MHC Classe I/imunologia , Rejeição de Enxerto/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Linfonodos/imunologia , Camundongos , Camundongos Nus , Camundongos SCID , Dados de Sequência Molecular , Transplante de Neoplasias , Peptídeos/imunologia , Transfecção
19.
J Biol Chem ; 274(23): 16513-21, 1999 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-10347215

RESUMO

Interleukin-10 (IL-10) is a cytokine that has pleiotropic effects on a variety of different cell types. Although many of the biologic responses induced by IL-10 are also induced by other cytokines, such as IL-6, IL-10 is relatively unique in its ability to potently inhibit production of pro-inflammatory cytokines in macrophages. In this study, we have used gain-of-function and loss-of-function genetic approaches to define the intracellular components involved in the different biologic actions of IL-10. Herein, we demonstrate that the ability of IL-10 to inhibit tumor necrosis factor alpha (TNFalpha) production in lipopolysaccharide-stimulated macrophages requires the presence of Stat3, Jak1, and two distinct regions of the IL-10 receptor intracellular domain. Macrophages deficient in Stat3 or Jak1 were unable to inhibit lipopolysaccharide-induced TNFalpha production following treatment with murine IL-10. Structure-function analysis of the intracellular domain of the IL-10 receptor alpha chain showed that whereas two redundant Stat3 recruitment sites (427YQKQ430 and 477YLKQ480) were required for all IL-10-dependent effects on either B cells or macrophages, expression of IL-10-dependent anti-inflammatory function required the presence on the intracellular domain of the IL-10 receptor of a carboxyl-terminal sequence containing at least one functionally critical serine. These results thus demonstrate that IL-10-induced inhibition of TNFalpha production requires two distinct regions of the IL-10 receptor intracellular domain and thereby establish a distinctive molecular basis for the developmental versus the anti-inflammatory actions of IL-10.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Inflamação/metabolismo , Proteínas Quinases Ativadas por Mitógeno , Receptores de Interleucina/fisiologia , Transdução de Sinais , Transativadores/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Animais , Linfócitos B/metabolismo , Feminino , Proteínas Quinases JNK Ativadas por Mitógeno , Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Interleucina-10 , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição STAT3 , Tirosina/metabolismo
20.
Nat Genet ; 21(4): 370-8, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10192386

RESUMO

The immunogenetic basis of severe infections caused by bacille Calmette-Guérin vaccine and environmental mycobacteria in humans remains largely unknown. We describe 18 patients from several generations of 12 unrelated families who were heterozygous for 1 to 5 overlapping IFNGR1 frameshift small deletions and a wild-type IFNGR1 allele. There were 12 independent mutation events at a single mutation site, defining a small deletion hotspot. Neighbouring sequence analysis favours a small deletion model of slipped mispairing events during replication. The mutant alleles encode cell-surface IFNgamma receptors that lack the intra-cytoplasmic domain, which, through a combination of impaired recycling, abrogated signalling and normal binding to IFNgamma exert a dominant-negative effect. We thus report a hotspot for human IFNGR1 small deletions that confer dominant susceptibility to infections caused by poorly virulent mycobacteria.


Assuntos
Predisposição Genética para Doença/genética , Infecções por Mycobacterium/imunologia , Receptores de Interferon/genética , Deleção de Sequência , Adolescente , Adulto , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Vacina BCG/efeitos adversos , Vacina BCG/uso terapêutico , Proteínas de Ligação a DNA/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Expressão Gênica , Predisposição Genética para Doença/imunologia , Heterozigoto , Humanos , Interferon gama/farmacologia , Masculino , Mycobacterium/patogenicidade , Infecções por Mycobacterium/genética , Linhagem , RNA Mensageiro/metabolismo , Receptores de Interferon/metabolismo , Fator de Transcrição STAT1 , Transativadores/metabolismo , Transfecção , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...