Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 11(10): 936-44, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20729857

RESUMO

Polarization of macrophages to M1 or M2 cells is important for mounting responses against bacterial and helminth infections, respectively. Jumonji domain containing-3 (Jmjd3), a histone 3 Lys27 (H3K27) demethylase, has been implicated in the activation of macrophages. Here we show that Jmjd3 is essential for M2 macrophage polarization in response to helminth infection and chitin, though Jmjd3 is dispensable for M1 responses. Furthermore, Jmjd3 (also known as Kdm6b) is essential for proper bone marrow macrophage differentiation, and this function depends on demethylase activity of Jmjd3. Jmjd3 deficiency affected trimethylation of H3K27 in only a limited number of genes. Among them, we identified Irf4 as encoding a key transcription factor that controls M2 macrophage polarization. Collectively, these results show that Jmjd3-mediated H3K27 demethylation is crucial for regulating M2 macrophage development leading to anti-helminth host responses.


Assuntos
Fatores Reguladores de Interferon/imunologia , Histona Desmetilases com o Domínio Jumonji/imunologia , Ativação de Macrófagos/imunologia , Macrófagos Alveolares/imunologia , Macrófagos/imunologia , Nippostrongylus/imunologia , Infecções por Strongylida/imunologia , Animais , Diferenciação Celular , Polaridade Celular , Quitina/imunologia , Regulação Enzimológica da Expressão Gênica , Histona Desmetilases/metabolismo , Interações Hospedeiro-Parasita/imunologia , Fatores Reguladores de Interferon/genética , Histona Desmetilases com o Domínio Jumonji/genética , Macrófagos/citologia , Metilação , Camundongos , Camundongos Knockout
2.
Eur J Immunol ; 48(8): 1319-1328, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29745988

RESUMO

Interferon regulatory factor 4 (IRF4) has critical roles in immune cell differentiation and function and is indispensable for clonal expansion and effector function in T cells. Here, we demonstrate that the AKT pathway is impaired in murine CD8+ T cells lacking IRF4. The expression of phosphatase and tensin homolog (PTEN), a negative regulator of the AKT pathway, was elevated in Irf4-/- CD8+ T cells. Inhibition of PTEN partially rescued downstream events, suggesting that PTEN constitutes a checkpoint in the IRF4-mediated regulation of cell signaling. Despite the clonal expansion defect, in the absence of IRF4, memory-like CD8+ T cells could be generated and maintained, although unable to expand in recall responses. The homeostatic proliferation of naïve Irf4-/- CD8+ T cells was impaired, whereas their number eventually reached a level similar to that of wild-type CD8+ T cells. Conversely, memory-like Irf4-/- CD8+ T cells underwent homeostatic proliferation in a manner similar to that of wild-type memory CD8+ T cells. These results suggest that IRF4 regulates the clonal expansion of CD8+ T cells at least in part via the AKT signaling pathway. Moreover, IRF4 regulates the homeostatic proliferation of naïve CD8+ T cells, whereas the maintenance of memory CD8+ T cells is IRF4-independent.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Fatores Reguladores de Interferon/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Diferenciação Celular/imunologia , Proliferação de Células , Células Cultivadas , Fatores Reguladores de Interferon/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PTEN Fosfo-Hidrolase/antagonistas & inibidores , Transdução de Sinais/imunologia
3.
Biochem Biophys Res Commun ; 501(4): 833-837, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29684346

RESUMO

Interferon regulatory factor (IRF) 4 and the proto-oncogene c-Rel cooperate in growth and antiviral drug resistance of adult T-cell leukemia/lymphoma (ATLL). To elucidate the target of IRF4 and c-Rel in ATLL, we determined the simultaneous binding sites of IRF4 and c-Rel using ChIP-seq technology. Nine genes were identified within 2 kb of binding sites, including MIR3662. Expression of miR-3662 was regulated by IRF4, and to a lesser extent by c-Rel. Cell proliferation was inhibited by knockdown of miR-3662 and expression of miR-3662 was correlated with antiviral drug resistance in ATLL cell lines. Thus, miR-3662 represents a target for therapies against ATLL.


Assuntos
Farmacorresistência Viral/genética , Regulação Leucêmica da Expressão Gênica , Leucemia-Linfoma de Células T do Adulto/genética , Leucemia-Linfoma de Células T do Adulto/virologia , MicroRNAs/genética , Adulto , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Humanos , Fatores Reguladores de Interferon/metabolismo , Leucemia-Linfoma de Células T do Adulto/patologia , MicroRNAs/metabolismo , Ligação Proteica/genética , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-rel/metabolismo
4.
PLoS Pathog ; 12(1): e1005357, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26735137

RESUMO

Dengue virus (DENV) is one of the most important arthropod-borne pathogens that cause life-threatening diseases in humans. However, no vaccine or specific antiviral is available for dengue. As seen in other RNA viruses, the innate immune system plays a key role in controlling DENV infection and disease outcome. Although the interferon (IFN) response, which is central to host protective immunity, has been reported to limit DENV replication, the molecular details of how DENV infection is modulated by IFN treatment are elusive. In this study, by employing a gain-of-function screen using a type I IFN-treated cell-derived cDNA library, we identified a previously uncharacterized gene, C19orf66, as an IFN-stimulated gene (ISG) that inhibits DENV replication, which we named Repressor of yield of DENV (RyDEN). Overexpression and gene knockdown experiments revealed that expression of RyDEN confers resistance to all serotypes of DENV in human cells. RyDEN expression also limited the replication of hepatitis C virus, Kunjin virus, Chikungunya virus, herpes simplex virus type 1, and human adenovirus. Importantly, RyDEN was considered to be a crucial effector molecule in the IFN-mediated anti-DENV response. When affinity purification-mass spectrometry analysis was performed, RyDEN was revealed to form a complex with cellular mRNA-binding proteins, poly(A)-binding protein cytoplasmic 1 (PABPC1), and La motif-related protein 1 (LARP1). Interestingly, PABPC1 and LARP1 were found to be positive modulators of DENV replication. Since RyDEN influenced intracellular events on DENV replication and, suppression of protein synthesis from DENV-based reporter construct RNA was also observed in RyDEN-expressing cells, our data suggest that RyDEN is likely to interfere with the translation of DENV via interaction with viral RNA and cellular mRNA-binding proteins, resulting in the inhibition of virus replication in infected cells.


Assuntos
Vírus da Dengue/fisiologia , Dengue/imunologia , Interferons/imunologia , Proteínas Virais/genética , Replicação Viral/imunologia , Linhagem Celular , Vírus da Dengue/crescimento & desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , Immunoblotting , Imunoprecipitação , Espectrometria de Massas , Reação em Cadeia da Polimerase , Transfecção
5.
J Immunol ; 192(5): 2271-9, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24489086

RESUMO

IRF4 is a transcription factor from the IRF factor family that plays pivotal roles in the differentiation and function of T and B lymphocytes. Although IRF4 is also expressed in dendritic cells (DCs) and macrophages, its roles in these cells in vivo are not clearly understood. In this study, conditional knockout mice that lack IRF4 in DCs or macrophages were generated and infected with Leishmania major. Mice lacking DC expression of IRF4 showed reduced footpad swelling compared with C57BL/6 mice, whereas those lacking IRF4 in macrophages did not. Mice with IRF4-deficient DCs also showed reduced parasite burden, and their CD4(+) T cells produced higher levels of IFN-γ in response to L. major Ag. In the draining lymph nodes, the proportion of activated CD4(+) T cells in these mice was similar to that in the control, but the proportion of IFN-γ-producing cells was increased, suggesting a Th1 bias in the immune response. Moreover, the numbers of migrating Langerhans cells and other migratory DCs in the draining lymph nodes were reduced both before and postinfection in mice with IRF4 defects in DCs, but higher levels of IL-12 were observed in IRF4-deficient DCs. These results imply that IRF4 expression in DCs inhibits their ability to produce IL-12 while promoting their migratory behavior, thus regulating CD4(+) T cell responses against local infection with L. major.


Assuntos
Fatores Reguladores de Interferon/imunologia , Interleucina-12/imunologia , Células de Langerhans/imunologia , Leishmania major/metabolismo , Leishmaniose Cutânea/imunologia , Células Th1/imunologia , Animais , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Fatores Reguladores de Interferon/genética , Interferon gama/imunologia , Interleucina-12/genética , Células de Langerhans/patologia , Leishmaniose Cutânea/genética , Leishmaniose Cutânea/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Células Th1/patologia
6.
Diabetologia ; 58(11): 2606-14, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26271342

RESUMO

AIMS/HYPOTHESIS: Interferon regulatory factor (IRF)4 plays a critical role in lymphoid development and the regulation of immune responses. Genetic deletion of IRF4 has been shown to suppress autoimmune disease in several mouse models, but its role in autoimmune diabetes in NOD mice remains unknown. METHODS: To address the role of IRF4 in the pathogenesis of autoimmune diabetes in NOD mice, we generated IRF4-knockout NOD mice and investigated the impact of the genetic deletion of IRF4 on diabetes, insulitis and insulin autoantibody; the effector function of T cells in vivo and in vitro; and the proportion of dendritic cell subsets. RESULTS: Heterozygous IRF4-deficient NOD mice maintained the number and phenotype of T cells at levels similar to NOD mice. However, diabetes and autoantibody production were completely suppressed in both heterozygous and homozygous IRF4-deficient NOD mice. The level of insulitis was strongly suppressed in both heterozygous and homozygous IRF4-deficient mice, with minimal insulitis observed in heterozygous mice. An adoptive transfer study revealed that IRF4 deficiency conferred disease resistance in a gene-dose-dependent manner in recipient NOD/severe combined immunodeficiency mice. Furthermore, the proportion of migratory dendritic cells in lymph nodes was reduced in heterozygous and homozygous IRF4-deficient NOD mice in an IRF4 dose-dependent manner. These results suggest that the levels of IRF4 in T cells and dendritic cells are important for the pathogenesis of diabetes in NOD mice. CONCLUSIONS/INTERPRETATION: Haploinsufficiency of IRF4 halted disease development in NOD mice. Our findings suggest that an IRF4-targeted strategy might be useful for modulating autoimmunity in type 1 diabetes.


Assuntos
Autoanticorpos/sangue , Diabetes Mellitus Tipo 1/genética , Haploinsuficiência , Insulina/imunologia , Fatores Reguladores de Interferon/genética , Animais , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/imunologia , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Fatores de Proteção , Linfócitos T/imunologia
7.
J Immunol ; 191(5): 2360-71, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23913959

RESUMO

IL-9 is a pleiotropic cytokine that can regulate autoimmune and allergic responses. Th9 cells can develop from naive T cells or Th2 cells through stimulation by TGF-ß in vitro. In this study, we demonstrated that Smad2 and Smad3 are necessary for IL-9 production from T cells in an OVA-induced asthma model using T cell-specific Smad2- and Smad3-deficient mice. Smad2 and Smad3 were also redundantly essential for TGF-ß signaling to induce histone modifications for Il9 transcription. Although Smad2/3 was recruited to the Il9 promoter by TGF-ß stimulation, they are not sufficient to activate the Il9 promoter. By the screening the transcription factors, we found that IFN regulatory factor 4 (IRF4) was essential for the Smad2/3-mediated Il9 promoter activation. In addition, Smad2/3 physically interacted with IRF4, and Smad2/3 did not bind to the Il9 promoter and could not induce Th9 in IRF4-deficient T cells. Similarly, IRF4 could not stimulate Il9 transcription in the absence of Smad2/3, and TGF-ß enhanced IRF4 recruitment to the Il9 promoter in a Smad2/3-dependent manner. We propose that Smad2/3 and IRF4 cooperatively transactivate the Il9 promoter and play an important role in regulating allergic immune responses by inducing Th9 cells.


Assuntos
Fatores Reguladores de Interferon/imunologia , Interleucina-9/imunologia , Ativação Linfocitária/imunologia , Proteína Smad2/imunologia , Proteína Smad3/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Western Blotting , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Citometria de Fluxo , Hipersensibilidade/imunologia , Fatores Reguladores de Interferon/metabolismo , Interleucina-9/biossíntese , Interleucina-9/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Subpopulações de Linfócitos T/metabolismo , Ativação Transcricional
8.
Hepatogastroenterology ; 62(139): 624-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26897942

RESUMO

BACKGROUND/AIMS: Though serum p53 antibody has been widely used, it is mentioned that it is not related to clinical parameters of colorectal cancer (CRC) and has no prognostic significance in long-term follow-up. The aim of this study was to explore the possibility to increase the value of p53 antibody in combination with carcinoembryonic antigen (CEA) and CA19-9 for post operation follow up. METHODOLOGY: One hundred twenty-eight patients with primary CRC who underwent surgery were enrolled in this study. Serum p53 antibodies, CEA and CA19-9 were measured before and after the surgery and their impact on CRC staging patient survival. DNA was extracted from colorectal cancer tissue and KRAS mutation analysis was determined by direct sequencing. RESULTS: Thirty seven point five patients were positive for serum p53 antibodies before the operation. Total 67.2% patients were positive with any tumor markers, but it was only 3.9% that were positive with all markers. There was no association between KRAS mutation and p53 antibody level. CONCLUSIONS: The combined use of tumor markers can be an effective screening method for detection of colorectal cancer. If serum p53 antibody level continues to be positive for long after resection, the case should require subsequent intense follow-up.


Assuntos
Autoanticorpos/sangue , Biomarcadores Tumorais/sangue , Colectomia , Neoplasias Colorretais/cirurgia , Proteína Supressora de Tumor p53/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Antígeno CA-19-9/sangue , Antígeno Carcinoembrionário/sangue , Colectomia/efeitos adversos , Colectomia/mortalidade , Neoplasias Colorretais/sangue , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Detecção Precoce de Câncer/métodos , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Mutação , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Proteínas Proto-Oncogênicas p21(ras)/genética , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
9.
Biochem Biophys Res Commun ; 447(1): 216-22, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24721431

RESUMO

Xenotropic murine leukemia virus-related virus (XMRV) is a novel gammaretrovirus that was originally isolated from human prostate cancer. It is now believed that XMRV is not the etiologic agent of prostate cancer. An analysis of murine leukemia virus (MLV) infection in various human cell lines revealed that prostate cancer cell lines are preferentially infected by XMRV, and this suggested that XMRV infection may confer some sort of growth advantage to prostate cancer cell lines. To examine this hypothesis, androgen-dependent LNCaP cells were infected with XMRV and tested for changes in certain cell growth properties. We found that XMRV-infected LNCaP cells can proliferate in the absence of the androgen dihydrotestosterone. Moreover, androgen receptor expression is significantly reduced in XMRV-infected LNCaP cells. Such alterations were not observed in uninfected and amphotropic MLV-infected LNCaP cells. This finding explains why prostate cancer cell lines are preferentially infected with XMRV.


Assuntos
Androgênios/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias da Próstata/virologia , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/fisiologia , Antagonistas de Receptores de Andrógenos/farmacologia , Anilidas/farmacologia , Animais , Linhagem Celular Tumoral , Di-Hidrotestosterona/farmacologia , Humanos , Masculino , Camundongos , Nitrilas/farmacologia , Ratos , Receptores Androgênicos/biossíntese , Receptores Androgênicos/efeitos dos fármacos , Compostos de Tosil/farmacologia
10.
Virus Genes ; 48(3): 448-56, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24469466

RESUMO

Ecotropic murine leukemia viruses (Eco-MLVs) infect mouse and rat, but not other mammalian cells, and gain access for infection through binding the cationic amino acid transporter 1 (CAT1). Glycosylation of the rat and hamster CAT1s inhibits Eco-MLV infection, and treatment of rat and hamster cells with a glycosylation inhibitor, tunicamycin, enhances Eco-MLV infection. Although the mouse CAT1 is also glycosylated, it does not inhibit Eco-MLV infection. Comparison of amino acid sequences between the rat and mouse CAT1s shows amino acid insertions in the rat protein near the Eco-MLV-binding motif. In addition to the insertion present in the rat CAT1, the hamster CAT1 has additional amino acid insertions. In contrast, tunicamycin treatment of mink and human cells does not elevate the infection, because their CAT1s do not have the Eco-MLV-binding motif. To define the evolutionary pathway of the Eco-MLV receptor, we analyzed CAT1 sequences and susceptibility to Eco-MLV infection of other several murinae animals, including the southern vole (Microtus rossiaemeridionalis), large Japanese field mouse (Apodemus speciosus), and Eurasian harvest mouse (Micromys minutus). Eco-MLV infection was enhanced by tunicamycin in these cells, and their CAT1 sequences have the insertions like the hamster CAT1. Phylogenetic analysis of mammalian CAT1s suggested that the ancestral CAT1 does not have the Eco-MLV-binding motif, like the human CAT1, and the mouse CAT1 is thought to be generated by the amino acid deletions in the third extracellular loop of CAT1.


Assuntos
Transportador 1 de Aminoácidos Catiônicos/genética , Evolução Molecular , Vírus da Leucemia Murina/fisiologia , Muridae/genética , Receptores Virais/genética , Infecções por Retroviridae/virologia , Doenças dos Roedores/virologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Arvicolinae , Transportador 1 de Aminoácidos Catiônicos/química , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Linhagem Celular , Cricetinae , Gerbillinae , Humanos , Camundongos , Dados de Sequência Molecular , Muridae/classificação , Muridae/virologia , Filogenia , Ratos , Receptores Virais/química , Receptores Virais/metabolismo
12.
Proc Natl Acad Sci U S A ; 108(46): 18766-71, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22042864

RESUMO

Mice deficient for interferon regulatory factor (Irf)2 (Irf2(-/-) mice) exhibit immunological abnormalities and cannot survive lymphocytic choriomeningitis virus infection. The pancreas of these animals is highly inflamed, a phenotype replicated by treatment with poly(I:C), a synthetic double-stranded RNA. Trypsinogen5 mRNA was constitutively up-regulated about 1,000-fold in Irf2(-/-) mice compared with controls as assessed by quantitative RT-PCR. Further knockout of IFNα/ß receptor 1(Ifnar1) abolished poly(I:C)-induced pancreatitis but had no effect on the constitutive up-regulation of trypsinogen5 gene, indicating crucial type I IFN signaling to elicit the inflammation. Analysis of Ifnar1(-/-) mice confirmed type I IFN-dependent transcriptional activation of dsRNA-sensing pattern recognition receptor genes MDA5, RIG-I, and TLR3, which induced poly(I:C)-dependent cell death in acinar cells in the absence of IRF2. We speculate that Trypsin5, the trypsinogen5 gene product, leaking from dead acinar cells triggers a chain reaction leading to lethal pancreatitis in Irf2(-/-) mice because it is resistant to a major endogenous trypsin inhibitor, Spink3.


Assuntos
Fator Regulador 2 de Interferon/metabolismo , Pancreatite/metabolismo , RNA de Cadeia Dupla/metabolismo , Transcrição Gênica , Tripsinogênio/genética , Células Acinares/metabolismo , Animais , Catepsina B/metabolismo , Glicoproteínas/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Pancreatite/genética , Poli I-C/genética , Proteínas Secretadas pela Próstata/metabolismo , Inibidor da Tripsina Pancreática de Kazal , Inibidores da Tripsina/farmacologia
13.
Small GTPases ; 13(1): 162-182, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34180342

RESUMO

We recently identified a CD63-interacting protein to understand the role of CD63 in virion production of the human immunodeficiency virus type 1, and we have found that Rab3a forms a complex with CD63. In this study, we analysed the effect of Rab3a on virion production of the murine leukaemia virus (MLV), which is another member of the retrovirus family. We found that Rab3a silencing induced lysosomal degradation of the MLV Gag protein, and recovery of the Rab3a expression restored the level of the Gag protein through a complex formation of MLV Gag and Rab3a, indicating that Rab3a is required for MLV Gag protein expression. In contrast, CD63 silencing decreased the infectivity of released virions but had no effect on virion production, indicating that CD63 facilitates the infectivity of released MLV particles. Although Rab3a induced CD63 degradation in uninfected cells, the complex of MLV Gag and Rab3a suppressed the Rab3a-mediated CD63 degradation in MLV-infected cells. Finally, we found that the MLV Gag protein interacts with Rab3a to stabilize its own protein and CD63 that facilitates the infectivity of released MLV particles. Considering the involvement of Rab3a in lysosome trafficking to the plasma membrane, it may also induce cell surface transport of the MLV Gag protein.


Assuntos
Produtos do Gene gag , Vírus da Leucemia Murina , Camundongos , Animais , Humanos , Produtos do Gene gag/metabolismo , Vírus da Leucemia Murina/metabolismo , Vírion/metabolismo , Membrana Celular/metabolismo , Proteínas de Ligação ao GTP/metabolismo
14.
Cells ; 11(14)2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35883685

RESUMO

Gamma-interferon (γ-IFN) significantly inhibits infection by replication-defective viral vectors derived from the human immunodeficiency virus type 1 (HIV-1) or murine leukemia virus (MLV) but the underlying mechanism remains unclear. Previously we reported that knockdown of γ-IFN-inducible lysosomal thiolreductase (GILT) abrogates the antiviral activity of γ-IFN in TE671 cells but not in HeLa cells, suggesting that other γ-IFN-inducible host factors are involved in its antiviral activity in HeLa cells. We identified cellular factors, the expression of which are induced by γ-IFN in HeLa cells, using a microarray, and analyzed the effects of 11 γ-IFN-induced factors on retroviral vector infection. Our results showed that the exogenous expression of FAT10, IFI6, or IDO1 significantly inhibits both HIV-1- and MLV-based vector infections. The antiviral activity of γ-IFN was decreased in HeLa cells, in which the function of IDO1, IFI6, FAT10, and GILT were simultaneously inhibited. IDO1 is an enzyme that metabolizes an essential amino acid, tryptophan. However, IDO1 did not restrict retroviral vector infection in Atg3-silencing HeLa cells, in which autophagy did not occur. This study found that IDO1, IFI6, FAT10, and GILT are involved in the antiviral activity of γ-IFN, and IDO1 inhibits retroviral infection by inducing autophagy.


Assuntos
Infecções por HIV , HIV-1 , Infecções por Retroviridae , Antirretrovirais/farmacologia , Antivirais/farmacologia , Autofagia , Infecções por HIV/tratamento farmacológico , HIV-1/metabolismo , Células HeLa , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interferon gama/metabolismo , Interferon gama/farmacologia , Vírus da Leucemia Murina , Proteínas Mitocondriais , Oxirredutases atuantes sobre Doadores de Grupo Enxofre , Ubiquitinas/farmacologia
15.
Cell Tissue Res ; 346(1): 79-88, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21987217

RESUMO

The importance of TNF-α signals mediated by tumor necrosis factor receptor type 1 (TNFR1) in inflammation and fibrosis induced by carbon tetrachloride (CCl(4)), and in post-injury liver regeneration including a GFP/CCl(4) model developed as a liver repair model by bone marrow cell (BMC) infusion, was investigated. In mice in which TNFR1 was suppressed by antagonist administration or by knockout, liver fibrosis induced by CCl(4) was significantly decreased. In these mice, intrahepatic macrophage infiltration and TGF-ß1 expression were reduced and stellate cell activity was decreased; however, expression of MMP-9 was also decreased. With GFP-positive BMC (TNFR1 wild-type, WT) infusion in these mice, fibrosis proliferation, including host endogenous intrahepatic macrophage infiltration, TGF-ß1 expression and stellate cell activity, increased significantly. There was no significant increase of MMP-9 expression. In this study, TNFR1 in hosts had a promoting effect on CCl(4)-induced hepatotoxicity and fibrosis, whereas BMC infusion in TNFR1 knockout mice enhanced host-derived intrahepatic inflammation and fibrosis proliferation. These findings differed from those in WT recipient mice, in which improvement in inflammation and fibrosis with BMC infusion had previously been reported. TNFR1-mediated signaling might be important to induce the improvement of liver fibrosis by bone marrow cell infusion.


Assuntos
Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Cirrose Hepática/metabolismo , Cirrose Hepática/terapia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Animais , Células da Medula Óssea/patologia , Tetracloreto de Carbono/toxicidade , Intoxicação por Tetracloreto de Carbono/genética , Intoxicação por Tetracloreto de Carbono/metabolismo , Intoxicação por Tetracloreto de Carbono/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/genética , Cirrose Hepática/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Transplante Homólogo
16.
Cytokine ; 56(3): 564-72, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21890374

RESUMO

Interferon regulatory factor (IRF)-4 is a member of the IRF transcription factor family, whose expression is primarily restricted to lymphoid and myeloid cells. In T-cells, IRF-4 expression is induced by T-cell receptor (TCR) cross-linking or treatment with phorbol-12-myristate-13-acetate (PMA)/Ionomycin, and IRF-4 is thought to be a critical factor for various functions of T-cells. To elucidate the IRF-4 functions in human adult T-cell leukemia virus type 1 (HTLV-1)-infected T-cells, which constitutively express IRF-4, we isolated IRF-4-binding proteins from T-cells, using a tandem affinity purification (TAP)-mass spectrometry strategy. Fourteen proteins were identified in the IRF-4-binding complex, including endogenous IRF-4 and the nuclear factor-kappaB (NF-κB) family member, c-Rel. The specific association of IRF-4 with c-Rel was confirmed by immunoprecipitation experiments, and IRF-4 was shown to enhance the c-Rel-dependent binding and activation of the interleukin-4 (IL-4) promoter region. We also demonstrated that IL-2 production was also enhanced by exogenously-expressed IRF-4 and c-Rel in the presence of P/I, in T-cells, and that the optimal IL-2 and IL-4 productions in vivo was IRF-4-dependent using IRF-4-/- mice. These data provide molecular evidence to support the clinical observation that elevated expression of c-Rel and IRF-4 is associated with the prognosis in adult T-cell leukemia/lymphoma (ATLL) patients, and present possible targets for future gene therapy.


Assuntos
Regulação da Expressão Gênica , Fatores Reguladores de Interferon/metabolismo , Interleucina-2/genética , Interleucina-4/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-rel/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Humanos , Interleucina-2/biossíntese , Interleucina-4/biossíntese , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Proteínas Proto-Oncogênicas c-rel/química
17.
Proc Natl Acad Sci U S A ; 105(41): 15890-5, 2008 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-18836070

RESUMO

Interferon regulatory factor (IRF) 4 is a member of the IRF family of transcription factors and plays critical roles in the development of CD4(+) T cells into Th2 and Th17 cells. Using the infection model of Nippostrongyrus brasiliensis, we have confirmed the critical roles of IRF-4 in Th2 development in vivo by using IRF-4(-/-) BALB/c mice. However, naïve IRF-4(-/-)CD4(+) T cells produced Th2 cytokines, including IL-4, IL-5, and IL-10, but not IL-2 or IFN-gamma, at levels higher than wild-type BALB/c CD4(+) T cells in response to T cell receptor stimulation. In contrast, effector/memory IRF-4(-/-)CD4(+) T cells did not exhibit increased production of Th2 cytokines. Knockdown of IRF-4 expression by using small interfering RNA promoted IL-4 production in naïve CD4(+) T cells but inhibited it in effector/memory CD4(+) T cells. These results indicate that IRF-4 plays differential roles in the regulation of Th2 cytokine production in naïve CD4(+) T cells and effector/memory CD4(+) T cells. IRF-4 inhibits Th2 cytokine production in naïve CD4(+) T cells, whereas it promotes Th2 cytokine production in effector/memory CD4(+) T cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Citocinas/biossíntese , Regulação da Expressão Gênica/imunologia , Fatores Reguladores de Interferon/fisiologia , Animais , Memória Imunológica , Fatores Reguladores de Interferon/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Th2/imunologia
18.
Cell Death Differ ; 28(12): 3199-3213, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34663907

RESUMO

SARS-CoV-2 vaccinations have greatly reduced COVID-19 cases, but we must continue to develop our understanding of the nature of the disease and its effects on human immunity. Previously, we suggested that a dysregulated STAT3 pathway following SARS-Co-2 infection ultimately leads to PAI-1 activation and cascades of pathologies. The major COVID-19-associated metabolic risks (old age, hypertension, cardiovascular diseases, diabetes, and obesity) share high PAI-1 levels and could predispose certain groups to severe COVID-19 complications. In this review article, we describe the common metabolic profile that is shared between all of these high-risk groups and COVID-19. This profile not only involves high levels of PAI-1 and STAT3 as previously described, but also includes low levels of glutamine and NAD+, coupled with overproduction of hyaluronan (HA). SARS-CoV-2 infection exacerbates this metabolic imbalance and predisposes these patients to the severe pathophysiologies of COVID-19, including the involvement of NETs (neutrophil extracellular traps) and HA overproduction in the lung. While hyperinflammation due to proinflammatory cytokine overproduction has been frequently documented, it is recently recognized that the immune response is markedly suppressed in some cases by the expansion and activity of MDSCs (myeloid-derived suppressor cells) and FoxP3+ Tregs (regulatory T cells). The metabolomics profiles of severe COVID-19 patients and patients with advanced cancer are similar, and in high-risk patients, SARS-CoV-2 infection leads to aberrant STAT3 activation, which promotes a cancer-like metabolism. We propose that glutamine deficiency and overproduced HA is the central metabolic characteristic of COVID-19 and its high-risk groups. We suggest the usage of glutamine supplementation and the repurposing of cancer drugs to prevent the development of severe COVID-19 pneumonia.


Assuntos
COVID-19/fisiopatologia , Glutamina/deficiência , Animais , COVID-19/sangue , COVID-19/epidemiologia , Comorbidade , Glutamina/sangue , Humanos , Ácido Hialurônico/sangue , Metaboloma , Inibidor 1 de Ativador de Plasminogênio/sangue , Fatores de Risco , Índice de Gravidade de Doença
19.
Biochem Biophys Res Commun ; 397(2): 202-7, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20501320

RESUMO

We analyzed the subcellular distributions and gene structures of interferon regulatory factor 3 (IRF3) transcription factor in 50 cases of human primary lung cancer. The immunohistochemical analyses revealed substantially aberrant IRF3 expression specific to the cancer lesions (2 and 6 tumors with nuclear staining, and 4 and 5 tumors with negative staining, in adenocarcinoma and squamous cell carcinoma, respectively), while the morphologically normal region around the tumors exhibited only cytoplasmic staining. In addition, we determined the sequence of the entire IRF3 coding region, and found two novel variants with the amino acid changes (S(175)(AGC)-->R(175)(CGC) and A(208)(GCC)-->D(208)(GAC)). The R(175) variant was also detected in a morphologically normal region around the nuclear staining squamous cell carcinoma, and exhibited almost the same functions as the wild type IRF3. On the other hand, the D(208) variant, found in the negative staining squamous cell carcinoma cases, reduced the nuclear translocation in response to IkappaB kinase epsilon stimulation, as compared to the wild type IRF3, but the same variant was detected in the surrounding morphologically normal region. The aberrant expression of IRF3 and the novel D(208) variant may provide clues to elucidate the etiology of primary lung cancer.


Assuntos
Adenocarcinoma/metabolismo , Carcinoma de Células Escamosas/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/patologia , Linhagem Celular , Feminino , Células HeLa , Humanos , Fator Regulador 3 de Interferon/genética , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Transporte Proteico/efeitos da radiação , Raios X
20.
Mol Immunol ; 46(4): 613-21, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18962896

RESUMO

IL-10 is a multifunctional cytokine that plays a critical role in maintaining the balance between immunity and tolerance. Previously, we identified proximal regulatory elements and alterations of chromatin structure in the IL-10 gene loci of Th1 and Th2 cells. We have now characterized a crucial cis-regulatory element, CNS-9, located 9kb upstream of the transcription start site in IL-10 gene loci. The CNS-9 region is highly conserved in vertebrate genomes, and contains clustered NFAT and IRF binding motifs. In vitro binding of NFAT1 and IRF4 to the CNS-9 region was observed by EMSA. Furthermore, Th2-preferential in vivo binding of NFAT1 and IRF4 to the CNS-9 region was observed by ChIP. Cyclosporine A treatment on wild type Th2 cells or Th2 cells derived from NFAT1 knockout (NFAT1(-/-)) mice showed significantly reduced trans-activity of CNS-9. The Th2 subset-specific enhancer activity of CNS-9 was upregulated synergistically by NFAT1 and its partner IRF4. Mutations in the binding sites for NFAT1 and IRF4 abrogated its enhancer activity of CNS-9. Collectively, our results establish crucial roles for enhancer element CNS-9, and NFAT1 and IRF4 that bind to it, for IL-10 expression in differential T helper subsets.


Assuntos
Elementos Facilitadores Genéticos/genética , Fatores Reguladores de Interferon/genética , Interleucina-10/genética , Fatores de Transcrição NFATC/genética , Células Th2/imunologia , Ativação Transcricional , Animais , Linhagem Celular Tumoral , Ciclosporina/farmacologia , Humanos , Fatores Reguladores de Interferon/metabolismo , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição NFATC/metabolismo , Transdução de Sinais/genética , Células Th1/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA