Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 15(6)2023 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-37376652

RESUMO

Macrophages are critical in the pathogenesis of a diverse group of viral pathogens, both as targets of infection and for eliciting primary defense mechanisms. Our prior in vitro work identified that CD40 signaling in murine peritoneal macrophages protects against several RNA viruses by eliciting IL-12, which stimulates the production of interferon gamma (IFN-γ). Here, we examine the role of CD40 signaling in vivo. We show that CD40 signaling is a critical, but currently poorly appreciated, component of the innate immune response using two distinct infectious agents: mouse-adapted influenza A virus (IAV, PR8) and recombinant VSV encoding the Ebola virus glycoprotein (rVSV-EBOV GP). We find that stimulation of CD40 signaling decreases early IAV titers, whereas loss of CD40 elevated early titers and compromised lung function by day 3 of infection. Protection conferred by CD40 signaling against IAV is dependent on IFN-γ production, consistent with our in vitro studies. Using rVSV-EBOV GP that serves as a low-biocontainment model of filovirus infection, we demonstrate that macrophages are a CD40-expressing population critical for protection within the peritoneum and T-cells are the key source of CD40L (CD154). These experiments reveal the in vivo mechanisms by which CD40 signaling in macrophages regulates the early host responses to RNA virus infection and highlight how CD40 agonists currently under investigation for clinical use may function as a novel class of broad antiviral treatments.


Assuntos
Antígenos CD40 , Infecções por Vírus de RNA , Vírus de RNA , Animais , Camundongos , Antígenos CD40/metabolismo , Interferon gama , Macrófagos , Infecções por Vírus de RNA/imunologia
2.
Cancers (Basel) ; 14(20)2022 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-36291813

RESUMO

TNF receptor-associated factor 3 (TRAF3) is an adapter protein that inhibits many signals that promote B cell survival and activation. Mice with a B cell-specific TRAF3 deficiency and humans with a rare haploinsufficiency in TRAF3 have enhanced development of BCLs as they age. Loss-of-function mutations in TRAF3 are common in B cell malignancies. Recent studies show that pharmacological inhibition of the enzyme glycogen synthase kinase 3 (GSK3), which regulates cellular growth, survival, and metabolism, inhibits growth and survival of BCL-derived B cells. In this study, we found that TRAF3 and GSK3 associated in B cells. The relative levels of TRAF3 in BCL cell lines correlated positively with the ratio of inactive to total GSK3ß, and negatively correlated with susceptibility to GSK3 inhibition by the GSK3 inhibitory drug 9-ING-41, currently in clinical trials. Uniquely in BCLs with low TRAF3, GSK3 inhibition caused increased loss of the TRAF3-regulated, anti-apoptotic protein Mcl-1. GSK3 inhibition also blocked hyperresponsiveness to IL-6 receptor signaling in TRAF3-deficient BCL cells. Together, these results support the utility of 9-ING-41 as a treatment for BCL, and suggest that a decrease or loss of TRAF3 in BCLs could act as a biomarker for increased susceptibility to GSK3 inhibitor treatment.

3.
J Leukoc Biol ; 109(2): 309-325, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32441445

RESUMO

Many acute viral infections target tissue Mϕs, yet the mechanisms of Mϕ-mediated control of viruses are poorly understood. Here, we report that CD40 expressed by peritoneal Mϕs restricts early infection of a broad range of RNA viruses. Loss of CD40 expression enhanced virus replication as early as 12-24 h of infection and, conversely, stimulation of CD40 signaling with an agonistic Ab blocked infection. With peritoneal cell populations infected with the filovirus, wild-type (WT) Ebola virus (EBOV), or a BSL2 model virus, recombinant vesicular stomatitis virus encoding Ebola virus glycoprotein (rVSV/EBOV GP), we examined the mechanism conferring protection. Here, we demonstrate that restricted virus replication in Mϕs required CD154/CD40 interactions that stimulated IL-12 production through TRAF6-dependent signaling. In turn, IL-12 production resulted in IFN-γ production, which induced proinflammatory polarization of Mϕs, protecting the cells from infection. These CD40-dependent events protected mice against virus challenge. CD40-/- mice were exquisitely sensitive to intraperitoneal challenge with a dose of rVSV/EBOV GP that was sublethal to CD40+/+ mice, exhibiting viremia within 12 h of infection and rapidly succumbing to infection. This study identifies a previously unappreciated role for Mϕ-intrinsic CD40 signaling in controlling acute virus infection.


Assuntos
Antígenos CD40/metabolismo , Imunidade Inata , Macrófagos/imunologia , Macrófagos/virologia , Vírus de RNA/fisiologia , Transdução de Sinais , Viroses/imunologia , Replicação Viral/fisiologia , Doença Aguda , Animais , Ligante de CD40/metabolismo , Ebolavirus/fisiologia , Glicoproteínas/imunologia , Humanos , Interferon gama/metabolismo , Interleucina-12/biossíntese , Camundongos Endogâmicos C57BL , Modelos Biológicos , Peritônio/patologia , Peritônio/virologia , Fator 6 Associado a Receptor de TNF/metabolismo , Viroses/virologia
4.
Front Immunol ; 11: 606936, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33613527

RESUMO

Systemic lupus erythematosus (SLE) and other autoimmune diseases are propelled by immune dysregulation and pathogenic, disease-specific autoantibodies. Autoimmunity against the lupus autoantigen Sm is associated with cross-reactivity to Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA-1). Additionally, EBV latent membrane protein-1 (LMP1), initially noted for its oncogenic activity, is an aberrantly active functional mimic of the B cell co-stimulatory molecule CD40. Mice expressing a transgene (Tg) for the mCD40-LMP1 hybrid molecule (containing the cytoplasmic tail of LMP1) have mild autoantibody production and other features of immune dysregulation by 2-3 months of age, but no overt autoimmune disease. This study evaluates whether exposure to the EBV molecular mimic, EBNA-1, stimulates antigen-specific and concurrently-reactive humoral and cellular immunity, as well as lupus-like features. After immunization with EBNA-1, mCD40-LMP1 Tg mice exhibited enhanced, antigen-specific, cellular and humoral responses compared to immunized WT congenic mice. EBNA-1 specific proliferative and inflammatory cytokine responses, including IL-17 and IFN-γ, were significantly increased (p<0.0001) in mCD40-LMP1 Tg mice, as well as antibody responses to amino- and carboxy-domains of EBNA-1. Of particular interest was the ability of mCD40-LMP1 to drive EBNA-1 associated molecular mimicry with the lupus-associated autoantigen, Sm. EBNA-1 immunized mCD40-LMP1 Tg mice exhibited enhanced proliferative and cytokine cellular responses (p<0.0001) to the EBNA-1 homologous epitope PPPGRRP and the Sm B/B' cross-reactive sequence PPPGMRPP. When immunized with the SLE autoantigen Sm, mCD40-LMP1 Tg mice again exhibited enhanced cellular and humoral immune responses to both Sm and EBNA-1. Cellular immune dysregulation with EBNA-1 immunization in mCD40-LMP1 Tg mice was accompanied by enhanced splenomegaly, increased serum blood urea nitrogen (BUN) and creatinine levels, and elevated anti-dsDNA and antinuclear antibody (ANA) levels (p<0.0001 compared to mCD40 WT mice). However, no evidence of immune-complex glomerulonephritis pathology was noted, suggesting that a combination of EBV and genetic factors may be required to drive lupus-associated renal disease. These data support that the expression of LMP1 in the context of EBNA-1 may interact to increase immune dysregulation that leads to pathogenic, autoantigen-specific lupus inflammation.


Assuntos
Autoantígenos/imunologia , Autoimunidade , Antígenos Nucleares do Vírus Epstein-Barr/imunologia , Imunidade Celular , Imunidade Humoral , Lúpus Eritematoso Sistêmico/imunologia , Mimetismo Molecular , Proteínas da Matriz Viral/imunologia , Proteínas Centrais de snRNP/imunologia , Animais , Anticorpos Antinucleares/sangue , Autoantígenos/administração & dosagem , Antígenos CD40/genética , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Reações Cruzadas , Epitopos , Antígenos Nucleares do Vírus Epstein-Barr/administração & dosagem , Imunização , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Proteínas Centrais de snRNP/administração & dosagem
5.
Sci Rep ; 9(1): 17502, 2019 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-31745166

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Sci Rep ; 9(1): 12884, 2019 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-31501481

RESUMO

TRAF3 is a versatile intracellular adapter protein with multiple context-specific roles. Uniquely in B cells, TRAF3 deficiency enhances survival and increases the risk of transformation, as loss of TRAF3 is observed in several types of B cell cancers. Here, we report a new mechanism for TRAF3 in the restraint of B cell survival. We found that TRAF3 deficiency was associated with induction of the pro-survival kinase Pim2 in mouse primary B cells and human malignant B cell lines. The increase in Pim2 was independent of NF-κB2 activation but was ameliorated with inhibition of STAT3 expression or function. TRAF3 deficiency also led to a Pim2-dependent increase in c-Myc protein levels and was associated with reduced c-Myc ubiquitination. TRAF3-deficient primary B cells were less sensitive to cell death induced by the Pim inhibitors SGI-1776 and TP-3654. Interestingly, human malignant B cell lines with low expression of TRAF3 were more sensitive to Pim inhibition-induced cell death. Combination treatment of TRAF3-deficient B cells and B cell tumor lines with c-Myc inhibitors enhanced their sensitivity to Pim inhibition, suggesting a possible therapeutic strategy. TRAF3 thus suppresses a Pim2-mediated B cell survival axis, which can be a potential target for treatment of B cell malignancies.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo , Animais , Linfócitos B/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Camundongos , Fosforilação , Fator de Transcrição STAT3/metabolismo , Fator 3 Associado a Receptor de TNF/deficiência
7.
Front Immunol ; 9: 2161, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30319624

RESUMO

The adaptor protein TNF receptor-associated factor 3 (TRAF3) serves as a powerful negative regulator in multiple aspects of B cell biology. Early in vitro studies in transformed cell lines suggested the potential of TRAF3 to inhibit signaling by its first identified binding receptor, CD40. However, because the canonical TRAF3 binding site on many receptors also mediates binding of other TRAFs, and whole-mouse TRAF3 deficiency is neonatally lethal, an accurate understanding of TRAF3's specific functions was delayed until conditional TRAF3-deficient mice were produced. Studies of B cell-specific TRAF3-deficient mice, complemented by investigations in normal and malignant mouse and human B cells, reveal that TRAF3 has powerful regulatory roles that are unique to this TRAF, as well as functions context-specific to the B cell. This review summarizes the current state of knowledge of these roles and functions. These include inhibition of signaling by plasma membrane receptors, negative regulation of intracellular receptors, and restraint of cytoplasmic NF- κB pathways. TRAF3 is also now known to function as a resident nuclear protein, and to impact B cell metabolism. Through these and additional mechanisms TRAF3 exerts powerful restraint upon B cell survival and activation. It is thus perhaps not surprising that TRAF3 has been revealed as an important tumor suppressor in B cells. The many and varied functions of TRAF3 in B cells, and new directions to pursue in future studies, are summarized and discussed here.


Assuntos
Linfócitos B/imunologia , Ativação Linfocitária , Transdução de Sinais/imunologia , Fator 3 Associado a Receptor de TNF/imunologia , Proteínas Supressoras de Tumor/imunologia , Animais , Sobrevivência Celular/imunologia , Humanos , Camundongos , Camundongos Knockout , NF-kappa B/imunologia
8.
Blood Adv ; 1(27): 2712-2723, 2017 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-29296923

RESUMO

Loss-of-function mutations in genes encoding the signaling protein tumor necrosis factor receptor-associated factor 3 (TRAF3) are commonly found in human B-cell malignancies, especially multiple myeloma and B-cell lymphoma (BCL). B-cell TRAF3 deficiency results in enhanced cell survival, elevated activation receptor signaling, and increased activity of certain transcriptional pathways regulating expression of prosurvival proteins. A recent analysis of TRAF3 protein staining of ∼300 human BCL tissue samples revealed that a higher proportion of samples expressing the oncogenic Epstein-Barr virus-encoded protein latent membrane protein 1 (LMP1) showed low/negative TRAF3 staining than predicted. LMP1, a dysregulated mimic of the CD40 receptor, binds TRAF3 more effectively than CD40. We hypothesized that LMP1 may sequester TRAF3, reducing its availability to inhibit prosurvival signaling pathways in the B cell. This hypothesis was addressed via 2 complementary approaches: (1) comparison of TRAF3-regulated activation and survival-related events with relative LMP1 expression in human BCL lines and (2) analysis of the impact upon such events in matched pairs of mouse BCL lines, both parental cells and subclones transfected with inducible LMP1, either wild-type LMP1 or a mutant LMP1 with defective TRAF3 binding. Results from both approaches showed that LMP1-expressing B cells display a phenotype highly similar to that of B cells lacking TRAF3 genes, indicating that LMP1 can render B cells functionally TRAF3 deficient without TRAF3 gene mutations, a finding of significant relevance to selecting pathway-targeted therapies for B-cell malignancies.

9.
Sci Rep ; 6: 35349, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27752131

RESUMO

The adaptor protein TNF receptor-associated factor 3 (TRAF3) is a critical regulator of B lymphocyte survival. B cell-specific TRAF3 deficiency results in enhanced viability and is associated with development of lymphoma and multiple myeloma. We show that TRAF3 deficiency led to induction of two proteins important for glucose metabolism, Glut1 and Hexokinase 2 (HXK2). This was associated with increased glucose uptake. In the absence of TRAF3, anaerobic glycolysis and oxidative phosphorylation were increased in B cells without changes in mitochondrial mass or reactive oxygen species. Chemical inhibition of glucose metabolism or glucose deprivation substantially attenuated the enhanced survival of TRAF3-deficient B cells, with a decrease in the pro-survival protein Mcl-1. Changes in Glut1 and Mcl-1 levels, glucose uptake and B cell number in the absence of TRAF3 were all dependent upon NF-κB inducing kinase (NIK). These results indicate that TRAF3 deficiency suffices to metabolically reprogram B cells, a finding that improves our understanding of the role of TRAF3 as a tumor suppressor, and suggests potential therapeutic strategies.


Assuntos
Linfócitos B/metabolismo , Linfoma/genética , Mieloma Múltiplo/genética , Fator 3 Associado a Receptor de TNF/genética , Animais , Reprogramação Celular/genética , Glucose/genética , Glucose/metabolismo , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Hexoquinase/genética , Hexoquinase/metabolismo , Linfoma/patologia , Camundongos Knockout , Mieloma Múltiplo/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator 3 Associado a Receptor de TNF/deficiência , Fator 3 Associado a Receptor de TNF/metabolismo , Quinase Induzida por NF-kappaB
10.
Proc Natl Acad Sci U S A ; 113(4): 1032-7, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26755589

RESUMO

The adaptor protein TNF receptor-associated factor 3 (TRAF3) regulates signaling through B-lymphocyte receptors, including CD40, BAFF receptor, and Toll-like receptors, and also plays a critical role inhibiting B-cell homoeostatic survival. Consistent with these findings, loss-of-function human TRAF3 mutations are common in B-cell cancers, particularly multiple myeloma and B-cell lymphoma. B cells of B-cell-specific TRAF3(-/-) mice (B-Traf3(-/-)) display remarkably enhanced survival compared with littermate control (WT) B cells. The mechanism for this abnormal homeostatic survival is poorly understood, a key knowledge gap in selecting optimal treatments for human B-cell cancers with TRAF3 deficiency. We show here for the first time to our knowledge that TRAF3 is a resident nuclear protein that associates with the transcriptional regulator cAMP response element binding protein (CREB) in both mouse and human B cells. The TRAF-C domain of TRAF3 was necessary and sufficient to localize TRAF3 to the nucleus via a functional nuclear localization signal. CREB protein was elevated in TRAF3(-/-) B cells, without change in mRNA, but with a decrease in CREB ubiquitination. CREB-mediated transcriptional activity was increased in TRAF3-deficient B cells. Consistent with these findings, Mcl-1, an antiapoptotic target of CREB-mediated transcription, was increased in the absence of TRAF3 and enhanced Mcl-1 was suppressed with CREB inhibition. TRAF3-deficient B cells were also preferentially sensitive to survival inhibition with pharmacologic CREB inhibitor. Our results identify a new mechanism by which nuclear TRAF3 regulates B-cell survival via inhibition of CREB stability, information highly relevant to the role of TRAF3 in B-cell malignancies.


Assuntos
Linfócitos B/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Proteínas Nucleares/fisiologia , Fator 3 Associado a Receptor de TNF/fisiologia , Adolescente , Adulto , Animais , Linhagem Celular , Sobrevivência Celular , Humanos , Linfoma de Células B/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Sinais de Localização Nuclear
11.
Sci Signal ; 8(392): ra88, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26329582

RESUMO

Tumor necrosis factor receptor-associated factor 3 (TRAF3) is an adaptor protein that inhibits signaling by CD40 and by the receptor for B cell-activating factor (BAFF) and negatively regulates homeostatic B cell survival. Loss-of-function mutations in TRAF3 are associated with human B cell malignancies, in particular multiple myeloma. The cytokine interleukin-6 (IL-6) supports the differentiation and survival of normal and neoplastic plasma cells. We found that mice with a deficiency in TRAF3 specifically in B cells (B-Traf3(-/-) mice) had about twice as many plasma cells as did their littermate controls. TRAF3-deficient B cells had enhanced responsiveness to IL-6, and genetic loss of IL-6 in B-Traf3(-/-) mice restored their plasma cell numbers to normal. TRAF3 inhibited IL-6 receptor (IL-6R)-mediated signaling by facilitating the association of PTPN22 (a nonreceptor protein tyrosine phosphatase) with the kinase Janus-activated kinase 1 (Jak1), which in turn blocked phosphorylation of the transcription factor STAT3 (signal transducer and activator of transcription 3). Consistent with these results, the number of plasma cells in the PTPN22-deficient mice was increased compared to that in the wild-type mice. Our findings identify TRAF3 and PTPN22 as inhibitors of IL-6R signaling in B cells and reveal a previously uncharacterized role for TRAF3 in the regulation of plasma cell differentiation.


Assuntos
Plasmócitos/imunologia , Receptores de Interleucina-6/imunologia , Transdução de Sinais/imunologia , Fator 3 Associado a Receptor de TNF/imunologia , Animais , Humanos , Camundongos , Camundongos Knockout , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética , Proteína Tirosina Fosfatase não Receptora Tipo 22/imunologia , Receptores de Interleucina-6/genética , Transdução de Sinais/genética , Fator 3 Associado a Receptor de TNF/genética
12.
Nat Immunol ; 15(9): 866-74, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25029551

RESUMO

The number of Foxp3+ regulatory T cells (Treg cells) must be tightly controlled for efficient suppression of autoimmunity with no impairment of normal immune responses. Here we found that the adaptor TRAF3 was intrinsically required for restraining the lineage determination of thymic Treg cells. T cell-specific deficiency in TRAF3 resulted in a two- to threefold greater frequency of Treg cells, due to the more efficient transition of precursors of Treg cells into Foxp3+ Treg cells. TRAF3 dampened interleukin 2 (IL-2) signaling by facilitating recruitment of the tyrosine phosphatase TCPTP to the IL-2 receptor complex, which resulted in dephosphorylation of the signaling molecules Jak1 and Jak3 and negative regulation of signaling via Jak and the transcription factor STAT5. Our results identify a role for TRAF3 as an important negative regulator of signaling via the IL-2 receptor that affects the development of Treg cells.


Assuntos
Diferenciação Celular/imunologia , Interleucina-2/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Fator 3 Associado a Receptor de TNF/imunologia , Timo/citologia , Animais , Autoimunidade/imunologia , Fatores de Transcrição Forkhead/imunologia , Janus Quinase 1/imunologia , Janus Quinase 3/imunologia , Camundongos , Proteína Tirosina Fosfatase não Receptora Tipo 2/imunologia , Fator de Transcrição STAT5/imunologia
13.
PLoS One ; 9(7): e102120, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25010048

RESUMO

Our laboratory reported previously that TNF receptor associated factor 3 (TRAF3) is a positive regulator of TCR signaling and T cell function. In the current study, we present new findings that reveal differential roles for TRAF3 in the regulation of CD4+ and CD8(+) T cells. In response to TCR stimulation in vitro, TRAF3 has greater impact in CD4(+) T cells than in CD8+ T cells. However, T cell-specific TRAF3 deficient mice (CD4Cre TRAF3(fl°x)/(fl°x); T-TRAF3(-/-)) have a greater number of CD4(+)CD44(hi) effector/memory T cells than littermate control (LMC) mice, possibly due to an inefficient suppressive effect of TRAF3 deficient Foxp3+ regulatory T cells. In contrast, CD8(+)CD44(hi)CD62L(hi) central memory (Tcm) cells are markedly reduced in T-TRAF3(-/-) mice in comparison to LMC mice, although CD8(+)CD44(hi)CD62L(l°w) effector memory T (Tem) cells and naïve T cells (CD8(+)CD44(l°w)CD62L(hi)) do not show significant differences in number. Importantly, TRAF3-deficient Tcm cells exhibit defective homeostasis due to impaired IL-15 signaling. These results indicate that the involvement of TRAF3 in IL-15 mediated signaling to T cells plays a previously unappreciated and critical role in CD8(+) Tcm cell regulation and maintenance.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Homeostase/imunologia , Memória Imunológica , Fator 3 Associado a Receptor de TNF/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Feminino , Interleucina-15/metabolismo , Ativação Linfocitária/imunologia , Masculino , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Fator 3 Associado a Receptor de TNF/deficiência
14.
Crit Rev Immunol ; 34(3): 177-98, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24941072

RESUMO

The Epstein Barr Virus (EBV)-encoded latent membrane protein 1 (LMP1) exerts numerous impacts on the functions of B lymphocytes, the cell type in which EBV establishes long-term latent infections. LMP1 expression has been implicated in making important contributions to a variety of human malignancies, as well as to autoimmune diseases. EBV also infects other types of immune cells, as well as nasopharyngeal epithelium, and evidence suggests that LMP1 functions may differ among cell types. In this review, we focus upon LMP1 functions in B cells. A variety of in vitro and in vivo model systems have been used by numerous groups of investigators to probe the ways in which LMP1 alters B-cell biology and the molecular mechanisms by which it exerts these effects. Here, we present a current overview of LMP1-mediated signaling pathways and downstream functions in B cells, the in vivo outcomes of LMP1 expression in model systems and humans, and the associations between LMP1 and human disease.


Assuntos
Linfócitos B/metabolismo , Linfócitos B/virologia , Herpesvirus Humano 4/fisiologia , Interações Hospedeiro-Patógeno , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Animais , Doenças Autoimunes/etiologia , Linfócitos B/imunologia , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/imunologia , Regulação Viral da Expressão Gênica , Humanos , Linfoma/etiologia , Camundongos , Modelos Animais , Transdução de Sinais
15.
Leuk Lymphoma ; 55(12): 2917-23, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24605938

RESUMO

Epstein-Barr virus (EBV) is associated with aggressive B cell lymphomas (BCLs). Latent membrane protein 1 (LMP1) of EBV is an oncogenic protein required for EBV B cell transformation. However, LMP1 is a weak oncogene in mice. Mice expressing Myc inserted 5' of the Eµ enhancer (iMyc(Eµ)), mimicking the t(8;14) translocation of endemic Burkitt lymphoma, develop delayed onset BCLs. To investigate potential cooperation between LMP1 and oncogenic MYC, we produced mice expressing the LMP1 signaling domain via a hybrid CD40-LMP1 transgene (mCD40-LMP1), and the dysregulated MYC protein of aggressive EBV+ BCLs. mCD40-LMP1/iMyc(Eµ) mice trended toward earlier BCL onset. BCLs from mCD40-LMP1/iMyc(Eµ) mice expressed LMP1 and were transplantable into immunocompetent recipients. iMyc(Eµ) and mCD40-LMP1/iMyc(Eµ) mice developed BCLs with similar immunophenotypes. LMP1 signaling was intact in BCLs as shown by inducible interleukin-6. Additionally, LMP1 signaling to tumor cells induced the two isoforms of Pim1, a constitutively active prosurvival kinase implicated in lymphomagenesis.


Assuntos
Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Expressão Gênica , Genótipo , Imunofenotipagem , Interleucina-6/metabolismo , Linfoma de Células B/mortalidade , Linfoma de Células B/virologia , Camundongos , Camundongos Transgênicos , Fenótipo , Prognóstico , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Transdução de Sinais
16.
Diabetes ; 63(8): 2751-60, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24647739

RESUMO

Chronic inflammation in visceral adipose tissue is considered a key element for induction of insulin resistance in obesity. CD40 is required for efficient systemic adaptive immune responses and is implicated in various inflammatory conditions. However, its role in modulating immunity in the microanatomical niches of adipose tissue remains largely undefined. Here, we show that, in contrast to its well-documented costimulatory effects, CD40 regulates development of insulin resistance in a diet-induced obesity (DIO) mouse model by ameliorating local inflammation in adipose tissues. CD40 deficiency (CD40KO) resulted in greater body weight gain, more severe inflammation in epididymal adipose tissue (EAT), and aggravated insulin resistance in response to DIO. Interestingly, we found that CD40KO CD8(+) T lymphocytes were major contributors to exacerbated insulin resistance. Specifically, CD8(+) T cells in EAT of DIO CD40KO mice produced elevated chemokines and proinflammatory cytokines and were critical for macrophage recruitment. These results indicate that CD40 plays distinct roles in different tissues and, unexpectedly, plays an important role in maintaining immune homeostasis in EAT. Further study of how CD40 promotes maintenance of healthy metabolism could contribute to better understanding of and ability to therapeutically manipulate the increasing health problem of obesity and insulin resistance.


Assuntos
Tecido Adiposo/metabolismo , Antígenos CD40/metabolismo , Homeostase , Ração Animal , Animais , Antígenos CD40/genética , Linfócitos T CD8-Positivos/fisiologia , Citocinas/genética , Citocinas/metabolismo , Dieta Hiperlipídica , Epididimo , Inflamação/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo
17.
Cytokine Growth Factor Rev ; 25(2): 147-56, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24433987

RESUMO

TRAF3 is an adapter protein that serves and regulates the functions of several types of receptors, located both inside the cell and at the plasma membrane. These include members of the TNF receptor superfamily (TNFR-SF), toll-like receptors (TLR), and cytokine receptors. It has become increasingly evident that the roles and functions of TRAF3 are highly context-dependent. TRAF3 can serve distinct roles for different receptors in the same cell, and also has highly cell-type-dependent functions. This review focuses upon the current state of knowledge regarding how TRAF3 regulates the biology and effector functions of B and T lymphocytes, two major cell types of the adaptive immune response in which TRAF3 has markedly distinct roles.


Assuntos
Linfócitos B/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Linfócitos T/imunologia , Fator 3 Associado a Receptor de TNF/imunologia , Animais , Humanos , Camundongos , NF-kappa B/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Citocinas/imunologia , Transdução de Sinais/imunologia , Fator 3 Associado a Receptor de TNF/genética , Receptores Toll-Like/imunologia
18.
Int Immunol ; 26(3): 149-58, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24170780

RESUMO

EBV-encoded latent membrane protein 1 (LMP1) is critical for EBV-driven B-cell transformation and most EBV-associated malignancies and is also implicated in exacerbation of autoimmunity. LMP1 functionally mimics the TNFR superfamily member CD40, but LMP1-induced signals and downstream B-cell functions are amplified and sustained compared with those mediated by CD40. CD40 and LMP1 both depend upon TNFR-associated factor (TRAF) adaptor molecules to mediate signaling but use them differently. LMP1 is dependent upon TRAFs 3 and 5 to deliver B-cell activation signals, while CD40 predominantly uses TRAFs 2 and 6 for this purpose. Both LMP1 and CD40 functions in B cells require TRAF6, which physically associates with both receptors but via different binding sites. In B-cell CD40 signaling, TRAF6 is required for a particular subset of CD40-dependent immune functions in vivo. Inasmuch as CD40 and LMP1 use other TRAFs differentially, we predicted that TRAF6 is critical for a specific subset of LMP1 functions in vivo and that this subset will be overlapping but distinct from the TRAF6-requiring functions of CD40. This study tests this prediction using a B-cell-specific TRAF6-deficient mouse model. We found that B-cell TRAF6 is important for LMP1-mediated antibody and autoantibody production in mice, as well as germinal center formation, but not the secondary lymphoid organ enlargement that results from LMP1 transgenic expression. Results highlight differential TRAF6 requirements for specific B-cell functions by LMP1 versus CD40. These differences may make important contributions to the contrasts between normally regulated CD40 versus pathogenic LMP1-mediated signals.


Assuntos
Linfócitos B/fisiologia , Antígenos CD40/metabolismo , Infecções por Vírus Epstein-Barr/imunologia , Centro Germinativo/patologia , Neoplasias Hematológicas/imunologia , Herpesvirus Humano 4/imunologia , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas da Matriz Viral/metabolismo , Animais , Autoimunidade/imunologia , Antígenos CD40/genética , Células Cultivadas , Modelos Animais de Doenças , Infecções por Vírus Epstein-Barr/complicações , Centro Germinativo/fisiologia , Neoplasias Hematológicas/etiologia , Humanos , Hiperplasia/genética , Imunidade Humoral/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais/genética , Fator 6 Associado a Receptor de TNF/genética , Proteínas da Matriz Viral/genética
19.
Biochem Biophys Res Commun ; 436(4): 660-5, 2013 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-23764397

RESUMO

Piperlongumine (PL), isolated from the fruit of Long pepper, Piper longum, is a cancer-inhibiting compound that selectively kills tumor cells while sparing their normal counterparts. Here we evaluated the efficacy with which PL suppresses malignant B cells derived from a newly developed, double-transgenic mouse model of human endemic Burkitt lymphoma (BL), designated mCD40-LMP1/iMyc(Eµ). PL inhibited tumor cell proliferation in a concentration-dependent manner and induced apoptosis of neoplastic but not normal B cells. Treatment with PL resulted in downregulation of EBV-encoded LMP1, cellular Myc, constitutive NF-κB activity, and a host of LMP1-Myc-NF-κB-regulated target genes including Aurka, Bcat1, Bub1b, Ccnb1, Chek1, Fancd2, Tfrc and Xrcc6. Of note, p21(Cip1)-encoding Cdkn1a was suppressed independent of changes in Trp53 mRNA levels and p53 DNA-binding activity. Considering the central role of the LMP1-NF-κB-Myc axis in B-lineage neoplasia, these findings further our understanding of the mechanisms by which PL inhibits B-lymphoma and provide a preclinical rationale for the inclusion of PL in new interventions in blood cancers.


Assuntos
Dioxolanos/farmacologia , Genes myc , Linfoma de Células B/patologia , Proteínas da Matriz Viral/fisiologia , Animais , Linhagem Celular Tumoral , Linfoma de Células B/genética , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo
20.
J Exp Med ; 210(6): 1079-86, 2013 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-23650438

RESUMO

TCR signaling is a prerequisite for early stage development of invariant natural killer T (iNKT) cells, whereas IL-15 signaling is required for expansion and maturation at later stages. In this study, we show that TNF receptor associated factor 3 (TRAF3) plays a critical role in the transition between these two distinct signaling pathways and developmental stages. TRAF3-deficient iNKT cells in CD4(Cre)TRAF3(flox/flox) (T-TRAF3(-/-)) mice exhibit defective up-regulation of T-bet and CD122, two critical molecules for IL-15 signaling, and as a consequence, IL-15-mediated iNKT cell proliferation and survival are impaired. Consistently, development of iNKT cells in T-TRAF3(-/-) mice shows a major defect at developmental stages 2 and 3, but not stages 0 and 1. We further demonstrated that defective T-bet up-regulation occurring during the stage 1 to stage 2 transition results from reduced TCR signaling in TRAF3(-/-) iNKT cells. In addition, mature TRAF3(-/-) iNKT cells displayed defective cytokine responses upon TCR stimulation. Collectively, our results reveal that by modulating the relative strength of TCR signaling, TRAF3 is an important regulator of iNKT cell development and functions.


Assuntos
Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Fator 3 Associado a Receptor de TNF/imunologia , Fator 3 Associado a Receptor de TNF/metabolismo , Animais , Processos de Crescimento Celular/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Interleucina-15/genética , Interleucina-15/imunologia , Interleucina-15/metabolismo , Subunidade beta de Receptor de Interleucina-2/genética , Subunidade beta de Receptor de Interleucina-2/imunologia , Subunidade beta de Receptor de Interleucina-2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia , Proteínas com Domínio T/metabolismo , Fator 3 Associado a Receptor de TNF/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...