Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
G Ital Dermatol Venereol ; 155(5): 574-589, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32618444

ABSTRACT

This article provides an overview of the main drugs to treat autoinflammatory disorders focusing on the four emblematic diseases within this group which represent, to date, the vast majority of patients with monogenic SAID; i.e. familial Mediterranean fever, mevalonate kinase deficiency, TNF receptor 1 deficiency and cryopyrin-associated periodic syndrome. We will therefore resume the evolutionary risks of the four main IL-1 dependent SAID, there treatments and monitoring tools. After having exposed the general principles, we will detail specific guidelines for the management in everyday clinical practice of patients according to the four main pathologies based on both our expertise and international recommendations. We aim herein to guide practitioners in charge of patients with common SAID towards optimal follow-up with appropriate monitoring of anti-inflammatory drugs.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/drug therapy , Familial Mediterranean Fever/drug therapy , Mevalonate Kinase Deficiency/drug therapy , TNF Receptor-Associated Factor 1/deficiency , Deficiency Diseases/drug therapy , Humans
2.
Cell Death Dis ; 5: e1467, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25321474

ABSTRACT

Tumor necrosis factor receptor-associated factor 1 (TRAF1), an adapter in signal transduction, is involved in immunity and in apoptotic processes in various cell types. However, little is known about its function and the molecular mechanism of its activation during liver injury. This study tested the hypothesis that TRAF1 is a mediator of cell injury after hepatic ischemia/reperfusion injury (I/R). In a mouse hepatic I/R injury model, we found that TRAF1 expression was highly induced. TRAF1 deficiency was liver protective, whereas sustained TRAF1 overexpression aggravated liver injury in response to hepatic I/R injury. Mechanistic studies demonstrated that a deficiency of TRAF1 in cultured hepatocytes led to the inhibition of NF-κB-mediated inflammatory responses, suppression of the ASK/JNK pro-death pathway and promotion of cellular regeneration capacity. In contrast, the converse occurred in hepatocyte-specific TRAF1 transgenic mice. TRAF1 activated the ASK1/JNK pathway and promoted hepatic injury. Our study demonstrates that TRAF1 is a crucial early mediator of hepatic I/R injury and suggests that TRAF1 may be a potential gene therapy target for the treatment of liver injury.


Subject(s)
Liver/blood supply , Liver/pathology , Reperfusion Injury/metabolism , TNF Receptor-Associated Factor 1/metabolism , Animals , Cell Survival , Cells, Cultured , Gene Expression Regulation , Hepatocytes/enzymology , Hepatocytes/pathology , Inflammation/pathology , Liver/metabolism , Liver Regeneration , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , TNF Receptor-Associated Factor 1/deficiency , TNF Receptor-Associated Factor 1/genetics
3.
J Exp Med ; 209(1): 77-91, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22184633

ABSTRACT

The signaling adaptor TNFR-associated factor 1 (TRAF1) is specifically lost from virus-specific CD8 T cells during the chronic phase of infection with HIV in humans or lymphocytic choriomeningitis virus (LCMV) clone 13 in mice. In contrast, TRAF1 is maintained at higher levels in virus-specific T cells of HIV controllers or after acute LCMV infection. TRAF1 expression negatively correlates with programmed death 1 expression and HIV load and knockdown of TRAF1 in CD8 T cells from viral controllers results in decreased HIV suppression ex vivo. Consistent with the desensitization of the TRAF1-binding co-stimulatory receptor 4-1BB, 4-1BBL-deficient mice have defects in viral control early, but not late, in chronic infection. TGFß induces the posttranslational loss of TRAF1, whereas IL-7 restores TRAF1 levels. A combination treatment with IL-7 and agonist anti-4-1BB antibody at 3 wk after LCMV clone 13 infection expands T cells and reduces viral load in a TRAF1-dependent manner. Moreover, transfer of TRAF1(+) but not TRAF1(-) memory T cells at the chronic stage of infection reduces viral load. These findings identify TRAF1 as a potential biomarker of HIV-specific CD8 T cell fitness during the chronic phase of disease and a target for therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , Lymphocytic Choriomeningitis/immunology , TNF Receptor-Associated Factor 1/deficiency , 4-1BB Ligand/immunology , 4-1BB Ligand/metabolism , Adoptive Transfer , Animals , Antibodies/immunology , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Chloroquine/pharmacology , Chronic Disease , Down-Regulation/genetics , Gene Expression , HIV Infections/genetics , Humans , Immunologic Memory , Interleukin-7/pharmacology , Lymphocytic Choriomeningitis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 1/genetics , Transforming Growth Factor beta/metabolism , Viral Load/immunology
4.
J Immunol ; 180(12): 8093-101, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18523273

ABSTRACT

During an acute immune response, CD8 T cells undergo rapid expansion followed by a contraction phase during which the majority of activated T cells die, leaving a few survivors to persist as memory cells. The regulation of T cell survival is critical at each stage of this response. 4-1BB, a TNFR family member, has been implicated in prolonging the survival of activated and memory CD8 T cells; however, the precise mechanisms by which 4-1BB sustains T cell survival are incompletely understood. Upon aggregation on T cells, 4-1BB associates with two TNFR-associated factors (TRAF), TRAF1 and TRAF2. TRAF2 is essential for downstream signaling from 4-1BB; however, the role of TRAF1 in 4-1BB signaling has not been elucidated and there have been conflicting data as to whether TRAF1 provides a positive or a negative signal in T cells. In this study, we report that TRAF1 plays a critical role in survival signaling downstream of 4-1BB during CD8 T cell expansion in response to viral infection in vivo. Further analysis reveals that TRAF1-deficient cells are impaired in their ability to up-regulate the prosurvival Bcl-2 family member Bcl-x(L) and show increased levels of the proapoptotic Bcl-2 family member Bim following 4-1BB signaling. TRAF1-deficient CD8 T cells fail to activate ERK in response to 4-1BB ligation and inhibition of ERK signaling downstream of 4-1BB in wild-type cells leads to increased Bim levels. Thus, TRAF1 has a prosurvival effect in CD8 T cells via the 4-1BB-mediated up-regulation of Bcl-x(L) and ERK-dependent Bim down-modulation.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/physiology , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Extracellular Signal-Regulated MAP Kinases/physiology , MAP Kinase Signaling System/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/physiology , TNF Receptor-Associated Factor 1/physiology , Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology , Animals , Apoptosis/genetics , Bcl-2-Like Protein 11 , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/virology , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , TNF Receptor-Associated Factor 1/deficiency , TNF Receptor-Associated Factor 1/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/deficiency , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , bcl-X Protein/biosynthesis
5.
Proc Natl Acad Sci U S A ; 103(49): 18703-8, 2006 Dec 05.
Article in English | MEDLINE | ID: mdl-17116875

ABSTRACT

The mechanisms that allow the maintenance of immunological memory remain incompletely defined. Here we report that tumor necrosis factor receptor (TNFR)-associated factor (TRAF) 1, a protein recruited in response to several costimulatory TNFR family members, is required for maximal CD8 T cell responses to influenza virus in mice. Decreased recovery of CD8 T cells in vivo occurred under conditions where cell division was unimpaired. In vitro, TRAF1-deficient, antigen-activated T cells accumulated higher levels of the proapoptotic BH3-only family member Bim, particularly the most toxic isoform, Bim(S). In the presence of excess IL-15, memory phenotype T cells with similar surface phenotype and comparable levels of Bcl-2 family members could be generated from WT or TRAF1-deficient T cell receptor transgenic OT-I T cells. However, when the memory CD8 T cells were allowed to compete for survival signals in the absence of antigen in vivo, the TRAF1-deficient T cells showed decreased recovery compared with TRAF1-sufficient T cells. This defect in T cell recovery in vivo was alleviated by introduction of siRNA to down-modulate Bim in TRAF1-deficient memory T cells. These studies identify the TRAF1 signaling axis and Bim down-regulation as critical for CD8 memory T cell survival in vivo.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Membrane Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , TNF Receptor-Associated Factor 1/physiology , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , CD8-Positive T-Lymphocytes/cytology , Cell Survival/genetics , Cell Survival/immunology , Immunologic Memory/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , Signal Transduction/immunology , TNF Receptor-Associated Factor 1/deficiency , TNF Receptor-Associated Factor 1/genetics
6.
Eur J Immunol ; 36(9): 2535-43, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16897814

ABSTRACT

The activation molecule CD40 and the death receptor CD95/Fas play important roles in regulating B cells so that effective antimicrobial immunity occurs without autoimmunity. CD40 signaling increases CD95 expression, sensitizing cells to apoptosis, but sustained CD40 signals rescue B cells from CD95 killing. Here we describe a mechanism of early CD40-mediated rescue from CD95-induced apoptosis in B cells. Maximal rescue was achieved when CD40 signals were given within 1-2 h of initiating CD95 apoptosis. CD40 signaling did not block association of Fas-associated death domain-containing protein with CD95, but decreased CD95-induced activation of caspases 3 and 8. Rapid CD40 rescue did not require NF-kappaB activation and was independent of de novo protein synthesis, but was dependent upon active PI3 K. Signaling via a CD40 mutant that does not bind TNFR-associated factor (TRAF)1, TRAF2, and TRAF3 rescued B cells from CD95-induced apoptosis. TRAF1/2/3-independent rescue was confirmed in B cell lines made deficient in these TRAF molecules by gene targeting. In contrast, CD40 rescue was completely abrogated in TRAF6-deficient B cells, which showed reduced activation of Akt in response to CD40 engagement. These results reveal a new rapid mechanism to balance B cell activation and apoptosis.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , CD40 Antigens/immunology , Phosphatidylinositol 3-Kinases/immunology , Receptors, Tumor Necrosis Factor/immunology , fas Receptor/immunology , Animals , B-Lymphocytes/metabolism , CD40 Antigens/metabolism , Caspases/immunology , Cell Line , Enzyme Activation/immunology , Immunoblotting , Immunoprecipitation , Lymphocyte Activation/immunology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Tumor Necrosis Factor/metabolism , TNF Receptor-Associated Factor 1/deficiency , TNF Receptor-Associated Factor 1/immunology , TNF Receptor-Associated Factor 2/deficiency , TNF Receptor-Associated Factor 2/immunology , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/immunology , TNF Receptor-Associated Factor 6/deficiency , TNF Receptor-Associated Factor 6/immunology , fas Receptor/metabolism
7.
J Immunol ; 176(9): 5388-400, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16622006

ABSTRACT

TNFR-associated factor 1 (TRAF1) is unique among the TRAF family, lacking most zinc-binding features, and showing marked up-regulation following activation signals. However, the biological roles that TRAF1 plays in immune cell signaling have been elusive, with many reports assigning contradictory roles to TRAF1. The overlapping binding site for TRAFs 1, 2, and 3 on many TNFR superfamily molecules, together with the early lethality of mice deficient in TRAFs 2 and 3, has complicated the quest for a clear understanding of the functions of TRAF1. Using a new method for gene targeting by homologous recombination in somatic cells, we produced and studied signaling by CD40 and its viral oncogenic mimic, latent membrane protein 1 (LMP1) in mouse B cell lines lacking TRAF1, TRAF2, or both TRAFs. Results indicate that TRAFs 1 and 2 cooperate in CD40-mediated activation of the B cell lines, with a dual deficiency leading to a markedly greater loss of function than that of either TRAF alone. In the absence of TRAF1, an increased amount of TRAF2 was recruited to lipid rafts, and subsequently, more robust degradation of TRAF2 and TRAF3 was induced in response to CD40 signaling. In contrast, LMP1 did not require either TRAFs 1 or 2 to induce activation. Taken together, our findings indicate that TRAF1 and TRAF2 cooperate in CD40 but not LMP1 signaling and suggest that cellular levels of TRAF1 may play an important role in modulating the degradation of TRAF2 and TRAF3 in response to signals from the TNFR superfamily.


Subject(s)
CD40 Antigens/metabolism , Signal Transduction , TNF Receptor-Associated Factor 1/metabolism , TNF Receptor-Associated Factor 2/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line , Enzyme Activation , Gene Expression Regulation , Immunoglobulins/biosynthesis , Immunoglobulins/immunology , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Microdomains/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , TNF Receptor-Associated Factor 1/deficiency , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 2/deficiency , TNF Receptor-Associated Factor 2/genetics
8.
Int Immunol ; 18(1): 101-11, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16352630

ABSTRACT

We have previously reported that tumor necrosis factor receptor-associated factor 1 (TRAF1), an intracellular protein, which binds to a range of molecules, including tumor necrosis factor (TNF) receptor family members, regulates TNF-induced NF-kappaB and AP-1 signaling as well as TCR-triggered proliferative responses in T cells. In order to define the role of TRAF1 in Th cell differentiation, we analyzed the responses of TRAF1-/- T cells following TCR activation. Stimulation of TRAF1-/- T cells by antigen resulted in significantly increased expression of the Th2 cytokines (IL-4, IL-5 and IL-13) compared with wild-type (WT) controls. The Th2 bias of TRAF1-/- T cells is T lymphocyte intrinsic, since naive CD4+CD62L+ TRAF1-/- T cells activated with CD3/CD28 produced elevated levels of Th2 cytokines. Consistent with these observations in cultured T cells, TRAF1-/- T cells induced enhanced Th2 responses in vivo. Transfer of ovalbumin (OVA)-immune TRAF1-/- T cells into naive WT recipients conferred significantly more intense pulmonary inflammation and higher airway hyperresponsiveness following inhaled OVA challenge than did transfer of OVA-immune WT T cells. Biochemical analysis of TRAF1-/- T cells revealed that they have elevated nuclear expression of NFAT-interacting protein (NIP45), a Th2 cell-associated transcription factor known to potentiate NFATp-driven IL-4 expression. In further experiments, we demonstrated that TRAF1 associates with a fraction of NIP45 in the cytoplasm and prevents its translocation to the nucleus. Taken together these results suggest that TRAF1 may limit the induction of Th2 responses by decreasing NIP45 concentration to the nucleus and thereby down-regulating the expression of NIP45-dependent IL-4 gene transcription.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation/immunology , Hypersensitivity/immunology , Intracellular Signaling Peptides and Proteins/immunology , Nuclear Proteins/immunology , TNF Receptor-Associated Factor 1/immunology , Th2 Cells/immunology , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Animals , Cell Differentiation/genetics , Cell Nucleus/genetics , Cell Nucleus/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytoplasm/genetics , Cytoplasm/immunology , Gene Expression Regulation/genetics , Hypersensitivity/genetics , Inflammation/genetics , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Proteins/genetics , TNF Receptor-Associated Factor 1/deficiency , Th2 Cells/transplantation
9.
J Virol ; 79(10): 6377-91, 2005 May.
Article in English | MEDLINE | ID: mdl-15858021

ABSTRACT

CD4(+)- and CD8(+)-T-cell death is a frequent immunological dysfunction associated with the development of human AIDS. We studied a murine model of AIDS, the CD4C/HIV transgenic (Tg) mouse model, to assess the importance of the apoptotic pathway in human immunodeficiency virus type 1 (HIV-1) pathogenesis. In these Tg mice, Nef is the major determinant of the disease and is expressed in immature and mature CD4(+) T cells and in cells of the macrophage/myeloid lineage. We report here a novel AIDS-like phenotype: enhanced death, most likely by apoptosis (as assessed by 7-aminoactinomycin D and annexin V/propidium iodide staining), of Tg thymic and peripheral CD4(+) and CD8(+) T cells. The Tg CD4(+) and CD8(+) T cells were also more susceptible to cell death after activation in vitro in mixed lymph node (LN) cultures. However, activation-induced cell death was not higher in Tg than in non-Tg-purified CD4(+) T cells. In addition, expression of Fas and FasL, assessed by flow cytometry, was increased in CD4(+) and CD8(+) T cells from Tg mice compared to that of non-Tg littermates. Despite the enhanced expression of Fas and FasL on Tg CD4(+) and CD8(+) T cells, Fas (lpr/lpr) and FasL (gld/gld) mutant CD4C/HIV Tg mice developed an AIDS-like disease indistinguishable from lpr/+ and gld/+ CD4C/HIV Tg mice, including loss of CD4(+) T cells. Similarly, CD4C/HIV Tg mice homozygous for mutations of two other genes implicated in cell death (interleukin-1beta-converting enzyme [ICE], tumor necrosis factor receptor 1 [TNFR-1]) developed similar AIDS-like disease as their respective heterozygous controls. Moreover, the double-Tg mice from a cross between the Bcl2/Wehi25 and CD4C/HIV Tg mice showed no major protection against disease. These results represent genetic evidence for the dispensable role of Fas, FasL, ICE, and TNFR-1 on the development of both T-cell loss and organ disease of these Tg mice. They also provide compelling evidence on the lack of protection by Bcl2 against Tg CD4(+)-T-cell death. In view of the high resemblance between numerous phenotypes observed in the CD4C/HIV Tg mice and in human AIDS, our findings are likely to be relevant for the human disease.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Apoptosis , Caspase 1/deficiency , Genes, nef/physiology , HIV-1/genetics , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins/metabolism , T-Lymphocytes/physiology , TNF Receptor-Associated Factor 1/deficiency , fas Receptor/metabolism , Acquired Immunodeficiency Syndrome/genetics , Acquired Immunodeficiency Syndrome/pathology , Animals , Caspase 1/genetics , Disease Models, Animal , Edema/pathology , Fas Ligand Protein , Kidney/pathology , Lymph Nodes/pathology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2 , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 1/genetics , Thymus Gland/pathology , Weight Loss , fas Receptor/genetics
10.
Neuroscience ; 131(2): 283-92, 2005.
Article in English | MEDLINE | ID: mdl-15708473

ABSTRACT

The major histocompatibility complex (MHC) glycoproteins, MHC1 and MHC2, play a key role in the presentation of antigen and the development of the immune response. In the current study we examined the regulation of the MHC2 in the mouse brain after facial axotomy. The normal facial motor nucleus showed very few slender and elongated MHC2+ cells. Transection of the facial nerve led to a gradual but strong upregulation in the number of MHC2+ cells, beginning at day 2 and reaching a maximum 14 days after axotomy, correlated with the induction of mRNA for tumor necrosis factor (TNF) alpha, interleukin (IL) 1beta and interferon-gamma (IFNgamma) and a peak in neuronal cell death. In almost all cases, MHC2 immunoreactivity was restricted to perivascular macrophages that colocalized with vascular basement membrane laminin and macrophage IBA1-immunoreactivity, with no immunoreactivity on phagocytic microglia, astrocytes or invading T-cells. Heterologous transplantation and systemic injection of endotoxin or IFNgamma did not affect this perivascular MHC2 immunoreactivity, and transgenic deletion of the IL1 receptor type I, or TNF receptor type 1, also had no effect. However, the deletion of IFNgamma receptor subunit 1 caused a significant increase, and that of TNF receptor type 2 a strong reduction in the number of MHC2+ macrophages, pointing to a counter-regulatory role of IFNgamma and TNFalpha in the immune surveillance of the injured nervous system.


Subject(s)
Facial Nerve Injuries/metabolism , Genes, MHC Class II/physiology , Macrophages/metabolism , Receptors, Interferon/physiology , Receptors, Tumor Necrosis Factor/physiology , Animals , Axotomy/methods , Facial Nerve/metabolism , Facial Nerve Injuries/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , TNF Receptor-Associated Factor 1/deficiency , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/physiology , TNF Receptor-Associated Factor 2/deficiency , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/physiology , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...