Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell Commun Signal ; 18(1): 30, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32093731

ABSTRACT

BACKGROUND: Candida albicans is the most common opportunistic human fungal pathogen. The chemokine ligand CXCL1 plays a protective role in fungal infection through the recruitment of neutrophils. TRAF1 (tumor necrosis factor-associated factor 1) can be highly induced by proinflammatory stimuli such as LPS and TNF and has been implicated in septic shock. However, the role of TRAF1 in infection, especially fungal infection, remains elusive. Herein, we reveal that TRAF1 suppresses the antifungal immune response to Candida albicans intradermal infection through the regulation of CXCL1 induction and neutrophil recruitment. METHODS: A mouse model of C. albicans intradermal infection was established. The Traf1-/- mice and Traf1-/- immortalized human keratinocytes were generated. The p65 inhibitor triptolide, STAT1 inhibitor fludarabine, neutrophil-depletion antibody Ly6G, and neutralizing antibody for CXCL1 were utilized. The expression of proinflammatory cytokines and chemokines was assessed by real-time PCR and ELISA, and the activation of signaling molecules was analyzed by Western blotting. Hematoxylin and eosin staining and periodic acid Schiff staining were used for histology or fungal detection, respectively. The immunofluorescence and flow cytometry analyses were employed in the assessment of immune cell infiltration. Bone marrow transplantation and adoptive transfer experiments were conducted to establish a role for TRAF1 in the macrophage compartment in fungal skin infection. RESULTS: TRAF1-deficient mice demonstrated improved control of Candida albicans intradermal infection, and concomitant increase in neutrophil recruitment and reduction in fungal burden. The chemokine CXCL1 was upregulated in the TRAF1-deficient macrophages treated with heat-killed C. albicans. Mechanistically, TRAF1-deficient macrophages showed increased activation of transcription factor NFκB p65. The human CXCL8 was also highly induced in the TRAF1-deficient human keratinocytes upon TNF stimulation through decreasing the activation of transcription factor STAT1. TRAF1-deficient macrophages played a critical role in containing the C. albicans skin infection in vivo. CONCLUSION: TRAF1-deficient mice can better control fungal infection in the skin, a process attributable to the CXCL-neutrophil axis. Mechanistically, TRAF1 likely regulates CXCL1 expression in both macrophages and keratinocytes through the transcriptional factor NFκB and STAT1, respectively. Our finding offers new insight into the understanding of the immune regulatory mechanisms in host defense against C. albicans infection.


Subject(s)
Candidiasis, Cutaneous/immunology , Chemokine CXCL1/immunology , Neutrophils , Skin/immunology , TNF Receptor-Associated Factor 1/immunology , Animals , Bone Marrow Cells , Female , HEK293 Cells , HaCaT Cells , Humans , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Neutrophils/immunology , Skin/cytology , Skin/pathology
2.
J Immunol ; 202(8): 2482-2492, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30867239

ABSTRACT

The TNFR superfamily member 4-1BB is important in the establishment of tissue-resident memory T cells (Trm) in the lung tissue following influenza infection. Moreover, supraphysiological boosting of 4-1BB in the airways during the boost phase of a prime-boost immunization regimen increases the long-lived Trm population, correlating with increased protection against heterotypic challenge. However, little is known about how 4-1BB contributes to the establishment of the lung Trm population. In this study, we show that effects of 4-1BB on lung Trm accumulation are already apparent at the effector stage, suggesting that the major role of 4-1BB in Trm formation is to allow persistence of CD8 T effector cells in the lung as they transition to Trm. Using supraphysiological stimulation of 4-1BB in the boost phase of a prime-boost immunization, we show that the effect of 4-1BB on Trm generation requires local delivery of both Ag and costimulation, is inhibited by rapamycin treatment during secondary CD8 effector T cell expansion, and is dependent on the signaling adaptor TRAF1. The decrease in lung Trm following early rapamycin treatment is accompanied by increased circulating memory T cells, as well as fewer effectors, suggesting a role for mammalian target of rapamycin (mTOR) in the formation of Trm through effects on the accumulation of effector precursors. Taken together, these data point to an important role for 4-1BB, TRAF1, and mTOR in the persistence of CD8 effector T cells in the lung parenchyma, thereby allowing the transition to Trm.


Subject(s)
4-1BB Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Influenza A virus/immunology , Lung Diseases/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , TNF Receptor-Associated Factor 1/immunology , TOR Serine-Threonine Kinases/immunology , 4-1BB Ligand/genetics , Animals , CD8-Positive T-Lymphocytes/pathology , Lung/pathology , Lung/virology , Lung Diseases/genetics , Lung Diseases/pathology , Lung Diseases/virology , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , TNF Receptor-Associated Factor 1/genetics , TOR Serine-Threonine Kinases/genetics
3.
Biochem Biophys Res Commun ; 511(1): 49-56, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30760405

ABSTRACT

Acute lung injury (ALI) is served as a severe life-threatening disease. However, the pathogenesis that contributes to ALI has not been fully understood. Tumor necrosis factor receptor-associated factor 1 (TRAF1) interacts with multiple regulators, performing its diverse role in biological functions. However, the effects of TRAF1 on ALI remain unknown. In this study, we attempted to explore the role of TRAF1 in ALI progression. The findings suggested that TRAF1-knockout (KO) markedly attenuated LPS-induced severe mortality rate in murine animals. LPS-elicited histological alterations in pulmonary tissues were significantly alleviated by TRAF1-deletion. Additionally, TRAF1 knockout effectively attenuated lung injury, as evidenced by the reduced lung wet/dry (W/D) weight ratio, as well as decreased bronchoalveolar lavage fluid (BALF) protein levels and neutrophil infiltration. Meanwhile, TRAF1 deletion markedly lessened inflammation, oxidative stress and apoptosis in BALF and/or lung tissues. The levels of pro-inflammatory cytokines stimulated by LPS were down-regulated by TRAF1 ablation, along with the inactivation of nuclear factor κB (NF-κB). LPS-promoted reactive oxygen species (ROS) generation was decreased in TRAF1-KO mice, partly through the improvement of anti-oxidants. Apoptosis was also inhibited by TRAF1 deletion in lung tissues of LPS-challenged mice through the suppression of cleaved Caspase-3. Moreover, TRAF1 knockout significantly decreased c-Jun N-terminal kinase (JNK) activation and its down-streaming signal of c-Jun in pulmonary samples of LPS-induced mice. Importantly, the in vitro study suggested that promoting JNK activation markedly abrogated TRAF1 knockdown-attenuated inflammation, ROS production and apoptosis in LPS-exposed A549 cells. Therefore, our experimental results provided evidence that TRAF1 suppression effectively protected LPS-induced ALI against inflammation, oxidative stress and apoptosis through the suppression of JNK activity.


Subject(s)
Acute Lung Injury/immunology , JNK Mitogen-Activated Protein Kinases/immunology , Lipopolysaccharides/immunology , TNF Receptor-Associated Factor 1/immunology , Acute Lung Injury/pathology , Animals , Cell Line , Enzyme Activation , Humans , Inflammation/immunology , Inflammation/pathology , Male , Mice, Inbred C57BL
4.
Front Immunol ; 9: 2969, 2018.
Article in English | MEDLINE | ID: mdl-30619326

ABSTRACT

Tumor necrosis factor receptor (TNFR) associated factor 1 (TRAF1) is a signaling adaptor first identified as part of the TNFR2 signaling complex. TRAF1 plays a key role in pro-survival signaling downstream of TNFR superfamily members such as TNFR2, LMP1, 4-1BB, and CD40. Recent studies have uncovered another role for TRAF1, independent of its role in TNFR superfamily signaling, in negatively regulating Toll-like receptor and Nod-like receptor signaling, through sequestering the linear ubiquitin assembly complex, LUBAC. TRAF1 has diverse roles in human disease. TRAF1 is overexpressed in many B cell related cancers and single nucleotide polymorphisms (SNPs) in TRAF1 have been linked to non-Hodgkin's lymphoma. Genome wide association studies have identified an association between SNPs in the 5' untranslated region of the TRAF1 gene with increased incidence and severity of rheumatoid arthritis and other rheumatic diseases. The loss of TRAF1 from chronically stimulated CD8 T cells results in desensitization of the 4-1BB signaling pathway, thereby contributing to T cell exhaustion during chronic infection. These apparently opposing roles of TRAF1 as both a positive and negative regulator of immune signaling have led to some confusion in the literature. Here we review the role of TRAF1 as a positive and negative regulator in different signaling pathways. Then we discuss the role of TRAF1 in human disease, attempting to reconcile seemingly contradictory roles based on current knowledge of TRAF1 signaling and biology. We also discuss avenues for future research to further clarify the impact of TRAF1 in human disease.


Subject(s)
Arthritis, Rheumatoid/genetics , B-Lymphocytes/immunology , Lymphoma, Non-Hodgkin/genetics , Signal Transduction/immunology , TNF Receptor-Associated Factor 1/metabolism , 5' Untranslated Regions/genetics , Arthritis, Rheumatoid/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Genome-Wide Association Study , Humans , Lymphoma, Non-Hodgkin/immunology , Polymorphism, Single Nucleotide/immunology , Signal Transduction/genetics , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Ubiquitin/metabolism , Ubiquitination/immunology
5.
Immunopharmacol Immunotoxicol ; 39(1): 28-36, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28000518

ABSTRACT

Artemisinin, isolated from the Chinese plant Artemisia annua, has been used for many years to treat different forms of malarial parasites. In this study, we explored the anti-inflammatory activity of artemisinin and the underlying mechanism of this action. We demonstrated that the anti-inflammatory effects of artemisinin in TPA-induced skin inflammation in mice. Then the artemisinin significantly inhibited the expression of NF-κB reporter gene induced by TNF-α in a dose-dependent manner. Artemisinin also inhibited TNF-α induced phosphorylation and degradation of IκBα, p65 nuclear translocation. Artemisinin also has an impact on upstream signaling of IKK through the inhibition of expression of adaptor proteins, TNF receptor-associated factor 2 (TRAF2) and receptor interacting protein 1 (RIP1). Furthermore, pretreatment of cells with artemisinin prevented the TNF-α-induced expression of NF-κB target genes, such as anti-apoptosis (c-IAP1, Bcl-2, and FLIP), proliferation (COX-2, cyclinD1), invasion (MMP-9), angiogenesis (VEGF), and major inflammatory cytokines (TNF-α, iNOS, and MCP1). We also proved that artemisinin potentiated TNF-α-induced apoptosis. Moreover, artemisinin significantly impaired the ROS production and phosphorylation of p38 and ERK, but did not affect the phosphorylation of JNK. Taken together, artemisinin may be a potentially useful therapeutic agent for inflammatory-related diseases.


Subject(s)
Artemisinins/pharmacology , MAP Kinase Signaling System/drug effects , NF-kappa B/immunology , Animals , Cell Line , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/immunology , MAP Kinase Signaling System/immunology , Mice , Nuclear Pore Complex Proteins/immunology , RNA-Binding Proteins/immunology , TNF Receptor-Associated Factor 1/immunology , TNF Receptor-Associated Factor 2/immunology , Tumor Necrosis Factor-alpha/adverse effects , Tumor Necrosis Factor-alpha/pharmacology
6.
Hum Immunol ; 76(1): 19-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25500258

ABSTRACT

The tumor necrosis factor receptor-associated factor 1/complement C5 (TRAF1/C5) genes have been suggested as two candidate genes for conferring susceptibility to autoimmunity and inflammation. The aim of the present study was to investigate the association of single nucleotide polymorphisms (SNP) of TRAF1/C5 genes with the risk for aplastic anemia (AA). In this case-control study, the genotyping of TRAF1/C5 rs10818488 polymorphism was detected by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The frequencies of AA, AG and GG genotypes, and A and G alleles were 21.9%, 52.4%, 25.7%, 48.1% and 51.9%, respectively, in AA patients. There was no significant differences in terms of genotype and allele distributions between AA patients and healthy controls (P=0.687 and 0.955, respectively). Similar results were found between the two groups when stratified by the disease severity including very severe AA (vSAA), SAA and non-SAA (NSAA). Our results indicated that TRAF1/C5 rs10818488 polymorphism might not contribute to susceptibility to AA in a Chinese population.


Subject(s)
Anemia, Aplastic/genetics , Complement C5/genetics , Polymorphism, Single Nucleotide , TNF Receptor-Associated Factor 1/genetics , Adolescent , Adult , Aged , Alleles , Anemia, Aplastic/immunology , Anemia, Aplastic/pathology , Case-Control Studies , China , Female , Gene Expression , Gene Frequency , Humans , Male , Middle Aged , Risk , Severity of Illness Index , TNF Receptor-Associated Factor 1/immunology
7.
PLoS One ; 8(8): e72421, 2013.
Article in English | MEDLINE | ID: mdl-24058413

ABSTRACT

The release of endogenous molecules from the skin after injury has been proposed to influence inflammation. Recent studies have found that pro-inflammatory signals can be generated by damaged endogenous self-RNA, and this event is detected by TLR3. Conversely, release of endogenous fragments of hyaluronic acid (HA) after injury has been proposed to inhibit LPS induced inflammation driven by TLR4. In this study we investigated if HA oligomers could also influence inflammation mediated by TLR3. A tetramer form of HA (oligo-HA) was added to MH-S cells (mouse alveolar macrophage cell line) that were then activated by poly(I:C). ELISA analysis of culture supernatants showed that the presence of oligo-HA suppressed the poly(I:C) induced release of IL-6 and TNFα. IL-6 mRNA expression was also suppressed as measured by quantitative RT-PCR. To determine the mechanism of action for oligo-HA to inhibit poly(I:C), macrophages derived from wild-type (WT), Tlr2-/- or Tlr4-/- mice were treated with oligo-HA and poly(I:C). Similar to WT cells, Tlr2-/- macrophages were inhibited by oligo-HA and retained suppression of cytokine release. In contrast, Tlr4-/- macrophages lost the capacity to be suppressed by oligo-HA. An increase in Traf1 (TLR negative regulator) mRNA was observed after oligo-HA treatment of WT but not in Tlr4-/- macrophages, and oligo-HA did not suppress cytokine responsiveness in Traf1-/- macrophages. These results show that oligo-HA acts through TLR4 and TRAF1 to inhibit TLR3-dependent inflammation. This observation illustrates the complex immunomodulatory action of endogenous products released after injury.


Subject(s)
Cytokines/immunology , Hyaluronic Acid/immunology , Macrophages, Alveolar/immunology , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/immunology , Animals , Cell Line , Cytokines/genetics , Gene Deletion , Gene Expression , Hyaluronic Acid/chemistry , Macrophages/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Oligosaccharides/chemistry , Oligosaccharides/immunology , Poly I-C/immunology , TNF Receptor-Associated Factor 1/immunology , Toll-Like Receptor 4/genetics
8.
J Clin Immunol ; 33(4): 759-66, 2013 May.
Article in English | MEDLINE | ID: mdl-23354839

ABSTRACT

PURPOSE: Recently, genomewide association analysis has revealed that the Tumor Necrosis Factor Receptor-associated factor 1-Complement 5 (TRAF1-C5) containing locus on chromosome 9 was associated with an increased risk for RA. Studies in model systems suggested that either gain- or loss-of-function TRAF1 mutations have immune effects that could plausibly lead to or exacerbate the arthritis phenotype. KRN/I-A(g7) (KxB/N) is a genetic mouse model of inflammatory arthritis. We aimed to assess the impact of TRAF1 deficiency on KRN/I-A(g7) mice. METHODS: We have bred KRN/I-A(g7) mice onto a TRAF1-deficient background and followed cohorts for the spontaneous appearance of arthritis. We have also transferred KxB/N serum to B6.I-A(g7) TRAF1KO recipients. In addition, systemic autoimmunity was induced through cGVH by injecting bm12 splenocytes into TRAF1KO recipient mice. RESULTS: TRAF1-deficient KRN/I-A(g7) mice spontaneously developed severe, progressive arthritis, comparable to that seen in TRAF1-intact KRN/I-A(g7) mice. However, the anti-GPI antibody titer was significantly lower in the former group. Interestingly, the TRAF1KO mice that had background levels of anti-GPI antibodies still showed severe arthritis, although with a brief delay compared to TRAF1 sufficient mice. In addition, TRAF1KO mice were fully susceptible to passive, serum transfer experiments. In another model of autoimmunity, TRAF1KO had no effect on cGVH autoantibodies production; nor was the response to an exogenous antigen impaired. CONCLUSION: The pathogenesis of spontaneous KRN/I-A(g7) arthritis can largely proceed by TRAF1-independent pathways. The production of anti-GPI autoantibody, but not other autoantibody or antibody responses, was markedly impaired by TRAF1 deficiency. The spontaneous arthritis model in KRN mice appears to be much less antibody dependent than previously believed.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Autoimmune Diseases/immunology , TNF Receptor-Associated Factor 1/immunology , Animals , Antibody Formation/drug effects , Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , Autoantibodies/blood , Autoimmune Diseases/genetics , Glucose-6-Phosphate Isomerase/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Risk , TNF Receptor-Associated Factor 1/genetics
9.
Clin Dev Immunol ; 2012: 487521, 2012.
Article in English | MEDLINE | ID: mdl-23125866

ABSTRACT

BACKGROUND: Polymorphisms of TRAF1 (Tumor necrosis factor receptor-associated factor 1) are associated with rheumatoid arthritis (RA). Whether TRAF1 polymorphisms confer increased risk for primary biliary cirrhosis (PBC), an autoimmune liver disease which can co-exist with RA, is unknown. AIM OF THE STUDY: To assess the frequency of the RA-conferring susceptibility TRAF1 polymorphisms rs3761847 and rs2900180 in a cohort of PBC patients. The association of TRAF1 polymorphisms with clinical features and autoantibody markers was also analyzed. METHODS: We studied 179 PBC patients and 300 controls. Samples were genotyped for TRAF1 gene polymorphisms by real-time PCR. Autoantibodies were tested by ELISA. RESULTS: The frequency of rs3761847 and rs2900180 polymorphisms did not differ between patients and controls. Laboratory or clinical features were not associated with specific polymorphisms. Gp210 autoantibody titres were conspicuously higher among GG homozygotes of rs3761847 as compared with AA homozygotes (P = 0.02). In contrast, antichromatin titers were higher in AA compared to GG rs3761847 homozygotes (P = 0.04). Rheumatoid factor IgG titres were significantly higher in rs2900180 TT homozygotes than CC homozygotes (P = 0.02). CONCLUSIONS: TRAF1 polymorphisms occur with the similar frequency in PBC patients and in the general population, but their presence is probably involved in the regulation of specific PBC-related autoantibodies.


Subject(s)
Autoantibodies/immunology , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/immunology , Nuclear Pore Complex Proteins/immunology , TNF Receptor-Associated Factor 1/genetics , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Autoantibodies/blood , Case-Control Studies , Cohort Studies , Female , Genetic Predisposition to Disease , Genotype , Homozygote , Humans , Liver Cirrhosis, Biliary/blood , Male , Middle Aged , Nuclear Pore Complex Proteins/blood , Polymorphism, Genetic , Risk , TNF Receptor-Associated Factor 1/immunology , Young Adult
10.
J Biol Chem ; 287(27): 23010-9, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22570473

ABSTRACT

T cells lacking TRAF1 hyperproliferate in response to T cell receptor signaling but have impaired signaling downstream of specific TNFR family members such as 4-1BB. Here we resolve this paradox by showing that while TRAF1 is required for maximal activation of the classical NF-κB pathway downstream of 4-1BB in primary T cells, TRAF1 also restricts the constitutive activation of NIK in anti-CD3-activated T cells. Activation of the alternative NF-κB pathway is restricted in unstimulated cells by a cIAP1/2:TRAF2:TRAF3:NIK complex. Using knockdown of NIK by siRNA we show that in activated CD8 T cells TRAF1 is also involved in this process and that constitutive activation of the alternative NF-κB pathway is responsible for costimulation independent hyperproliferation and excess cytokine production in TRAF1-deficient CD8 T cells compared with WT CD8 T cells. The T cell costimulatory molecule 4-1BB critically regulates the survival of activated and memory CD8 T cells. We demonstrate that stimulation through 4-1BB induces cIAP1-dependent TRAF3 degradation and activation of the alternative NF-κB pathway. We also show that while both TRAF1 and cIAP1 have non-redundant roles in suppressing the alternative NF-κB pathway in T cells activated in the absence of costimulation, activation of the classical NF-κB pathway downstream of 4-1BB requires TRAF1, whereas cIAP1 plays a redundant role with cIAP2. Collectively these results demonstrate that TRAF1 plays a critical role in regulating T cell activation both through restricting the costimulation independent activation of NIK in activated T cells and by promoting the 4-1BB-induced classical NF-κB pathway.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , NF-kappa B/metabolism , Signal Transduction/immunology , TNF Receptor-Associated Factor 1/immunology , TNF Receptor-Associated Factor 1/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Division/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , TNF Receptor-Associated Factor 1/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , NF-kappaB-Inducing Kinase
11.
Cell Signal ; 24(6): 1297-305, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22374304

ABSTRACT

Tumor necrosis factor (TNF) is a key mediator in the inflammatory response which is implicated in the onset of a number of diseases. Research on TNF led to the characterization of the largest family of cytokines known until now, the TNF superfamily, which exert their biological effects through the interaction with transmembrane receptors of the TNFR superfamily. TNF itself exerts its biological effects interacting with two different receptors: TNFR1 and TNFR2. TNFR1 presents a death domain on its intracellular region. In contrast to TNFR1, TNFR2 does not have a death domain. Activation of TNFR1 implies the consecutive formation of two different TNF receptor signalling complexes. Complex I controls the expression of antiapoptotic proteins that prevent the triggering of cell death processes, whereas Complex II triggers cell death processes. TNFR2 only signals for antiapoptotic reactions. However, recent evidence indicates that TNFR2 also signals to induce TRAF2 degradation. TRAF2 is a key mediator in signal transduction of both TNFR1 and TNFR2. Thus, this novel signalling pathway has two important implications: on one hand, it represents an auto regulatory loop for TNFR2; on the other hand, when this signal is triggered TNFR1 activity is modified so that antiapoptotic pathways are inhibited and apoptotic reactions are enhanced.


Subject(s)
Receptors, Tumor Necrosis Factor/immunology , Signal Transduction , Amino Acid Sequence , Animals , Apoptosis , Humans , Molecular Sequence Data , Receptors, Tumor Necrosis Factor/metabolism , Sequence Alignment , TNF Receptor-Associated Factor 1/chemistry , TNF Receptor-Associated Factor 1/immunology , TNF Receptor-Associated Factor 1/metabolism , TNF Receptor-Associated Factor 2/chemistry , TNF Receptor-Associated Factor 2/immunology , TNF Receptor-Associated Factor 2/metabolism
12.
Cell Immunol ; 273(1): 67-72, 2012.
Article in English | MEDLINE | ID: mdl-22196377

ABSTRACT

Rheumatoid arthritis (RA) is the most common cause of adult inflammatory arthritis. Recent genome-wide association scans have disclosed several single-nucleotide polymorphisms associated with RA susceptibility. The aim of this study was to determine whether the polymorphisms of TRAF1/C5 (tumor necrosis factor (TNF)-receptor associated factor 1)/(complement component 5) and STAT4 (signal transducers and activators of transcription 4) confer susceptibility, activity and severity to RA in Egyptian populations. One hundred and seventy-two RA patients and 160 controls were enrolled in the study. Polymorphisms of TRAF1/C5 and STAT4 genes were determined using restriction fragment length polymorphism-polymerase chain reaction. The TRAF1/C5 A and STAT4 T alleles were significantly associated with RA in Egyptian population. TRAF1/C5 A allele and STAT4 TT genotype were significantly associated with RA severity. In conclusion the mutant alleles or genotypes of both examined polymorphisms are associated with the development of RA in Egyptian population.


Subject(s)
Arthritis, Rheumatoid/immunology , STAT4 Transcription Factor/immunology , TNF Receptor-Associated Factor 1/immunology , Adult , Aged , Arthritis, Rheumatoid/genetics , Chi-Square Distribution , DNA/chemistry , DNA/genetics , Egypt , Female , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Middle Aged , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide , Rheumatoid Factor/blood , STAT4 Transcription Factor/genetics , TNF Receptor-Associated Factor 1/genetics
13.
Hum Immunol ; 72(12): 1210-3, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21968398

ABSTRACT

A significant source of variability in the literature on systemic lupus erythematosus (SLE) susceptibility genes has been the inability to replicate genetic findings across different racial or ethnic groups. We investigated whether a single nucleotide polymorphism (SNP) of the STAT4 (rs7574865), PTPN22 (rs2476601), TRAF1/C5 (rs10818488), and C1q (rs292001) genes as well as the 27-bp VNTR polymorphism on intron 4 of eNOS, previously associated with SLE in other populations, are also associated with SLE risk in Turkey. A group of 158 SLE patients and 155 healthy controls were included in this study. A genetic association of the TRAF1/C5, C1q, and eNOS gene polymorphism, but not of STAT4 and PTPN22, was found to confer a degree of risk for SLE. These data highlight the importance of comparative studies in different populations to confirm the previously detected genetic associations.


Subject(s)
Complement C1q/genetics , Genetic Predisposition to Disease , Lupus Erythematosus, Systemic/genetics , Nitric Oxide Synthase Type III/genetics , Polymorphism, Single Nucleotide/immunology , TNF Receptor-Associated Factor 1/genetics , Adult , Case-Control Studies , Cohort Studies , Complement C1q/immunology , Female , Genetic Association Studies , Genotype , Humans , Lupus Erythematosus, Systemic/epidemiology , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged , Nitric Oxide Synthase Type III/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Risk Factors , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/immunology , TNF Receptor-Associated Factor 1/immunology , Turkey/epidemiology
15.
Ann Rheum Dis ; 69(3): 561-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19401279

ABSTRACT

BACKGROUND: Genome-wide association studies have proposed susceptibility variants for rheumatoid arthritis in the TRAF1-C5 locus and 6q23 region. Furthermore, additional independent studies have investigated the same or highly linked polymorphisms in the same regions. OBJECTIVE: To carry out a meta-analysis of the available evidence for the association of polymorphisms in the TRAF1-C5 locus and 6q23 region with rheumatoid arthritis. METHODS: Data were synthesised for four polymorphisms: rs3761847 (n=13 datasets) and rs2900180 (n=9 datasets) in the TRAF1-C5 locus, and rs10499194 (n=5 datasets) and rs6920220 (n=7 datasets) in the 6q23 region. Meta-analyses for subgroups defined by anti-cyclic citrullinated peptide (anti-CCP) and rheumatoid factor (RF) status were also performed. RESULTS: The polymorphism rs6920220 reached genome-wide statistical significance with p=7.9 x 10(-17) and an allelic odds ratio of 1.24 (95% CI 1.18 to 1.30) and no between-study heterogeneity (I(2)=0%). The risk was significantly stronger in patients with anti-CCP antibodies and in patients with RF. The other three variants showed large between-study heterogeneity across datasets (I(2) range 74-82%); rs10499194 was nominally statistically significant after exclusion of the discovery data. Two variants had genome-wide statistical significance in subgroups defined by the presence of RF (rs3761847 and rs6920220) or anti-CCP (rs6920220). CONCLUSIONS: Genetic markers in the 6q23 region and TRAF1-C5 are associated with rheumatoid arthritis, in particular with positive anti-CCP and RF profile. With the exception of rs6920220, which shows highly consistent results, other proposed markers have high between-study heterogeneity that may reflect unrecognised phenotypic or genetic variability (eg, gene environment interactions) within rheumatoid arthritis. Furthermore, these markers may not be the true causative loci but rather be in linkage disequilibrium with the true ones.


Subject(s)
Arthritis, Rheumatoid/genetics , Chromosomes, Human, Pair 6/genetics , TNF Receptor-Associated Factor 1/genetics , Arthritis, Rheumatoid/immunology , Chromosome Mapping , Chromosomes, Human, Pair 6/immunology , Complement C5/genetics , Epidemiologic Methods , Genetic Loci , Genetic Markers , Genetic Predisposition to Disease , Humans , Peptides, Cyclic/genetics , Polymorphism, Genetic , Rheumatoid Factor/genetics , Rheumatoid Factor/immunology , TNF Receptor-Associated Factor 1/immunology
16.
Mol Immunol ; 46(16): 3278-82, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19698991

ABSTRACT

Tumor necrosis factor receptor-associated factor 1 (TRAF1) is unique among the members of the TRAF family, as it lacks the N-terminal RING/zinc-finger domain. Also the function of TRAF1 is not clearly established, with many papers reporting contradictory results. Here we show that TRAF1 interacts with BAFF receptor, a member of the TNF receptor family, and positively regulates activation of the alternative NF-kappaB pathway. Ectopic expression of TRAF1 causes degradation of TRAF3, stabilization of NIK, and processing of p100 to produce the mature form p52. In addition, we show that knocking-down expression of TRAF1 in the Hodgkin's disease derived cell line L1236, interfere with p100 processing and with p52 mediate gene transcription. Collectively these results support a role for TRAF1 as a positive regulator of the NF-kappaB alternative pathway.


Subject(s)
B-Cell Activation Factor Receptor/biosynthesis , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 1/biosynthesis , TNF Receptor-Associated Factor 3/metabolism , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/immunology , Cell Line, Tumor , Gene Expression , Hodgkin Disease/genetics , Hodgkin Disease/immunology , Hodgkin Disease/metabolism , Humans , NF-kappa B/genetics , NF-kappa B/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Stability , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/immunology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , Zinc Fingers/genetics , Zinc Fingers/immunology , NF-kappaB-Inducing Kinase
18.
Immunology ; 120(3): 303-14, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17328785

ABSTRACT

Inhaled lipopolysaccharide (LPS) induces an inflammatory response that may contribute to the pathogenesis of asthma and other airway diseases. Here we investigate the role of tumour necrosis factor (TNF) receptor-associated factor 1 (TRAF1) in leucocyte recruitment using a model of LPS-induced lung inflammation in mice. TRAF1(-/-) mice are completely deficient in the recruitment of lymphocytes to the lower respiratory tract after inhalation of LPS. Although TRAF1(-/-) mice display normal early accumulation of neutrophils, dendritic cells and monocytes in the alveolar airspace, they have a significantly reduced recruitment of these cells by 24 hr after inhalation of LPS when compared to wild-type (WT) mice. Despite normal expression of the pro-inflammatory cytokines TNF, interleukin-1 (IL-1) and IL-6 after LPS treatment, TRAF1(-/-) mice displayed decreased expression of intercellular adhesion molecule 1, vascular cell adhesion molecule 1, CCL17 and CCL20 in the lungs, when compared to LPS-treated WT mice. These results suggest that TRAF1 facilitates LPS-induced leucocyte recruitment into the lung airways by augmenting the expression of chemokines and adhesion molecules. Mice lacking TNF receptor 1 (TNFR1) but not TNFR2 show a phenotype similar to the TRAF1(-/-) mice, suggesting that TRAF1 may act downstream of TNFR1. Significantly, we use bone marrow chimeras to demonstrate that expression of TRAF1 by cells resident in the lungs, but not by circulating leucocytes, is necessary for efficient LPS-induced recruitment of leucocytes to the lung airways.


Subject(s)
Leukocytes/immunology , Lipopolysaccharides/immunology , Lung/immunology , Pneumonia/immunology , TNF Receptor-Associated Factor 1/immunology , Animals , Bone Marrow Cells/immunology , Bronchoalveolar Lavage Fluid/immunology , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Chemotaxis, Leukocyte/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Neutrophils/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/immunology
19.
J Invest Dermatol ; 127(8): 1898-904, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17392826

ABSTRACT

Lymphomatoid papulosis (LyP), primary cutaneous anaplastic large T-cell lymphoma (cALCL), and cutaneous infiltrates of systemic anaplastic large cell lymphoma (sALCL) are CD30-positive lymphoproliferative disorders of the skin that overlap clinically, histopathologically, immunophenotypically, and genetically but differ considerably in their prognosis. In particular, lesions of LyP regress spontaneously, whereas those of cALCL and sALCL persist and may progress and spread to extracutaneous sites. In contrast to patients with cALCL, LyP patients do not benefit from an aggressive radio- and/or chemotherapeutic approach. We generated a novel tumor necrosis factor receptor (TNFR)-associated factor 1 (TRAF1) antibody that recognizes a formalin-resistant epitope (Ber-TRAF1A) and investigated the expression of TRAF1, an intracellular component of TNFR signaling, in LyP and ALCL. We could show a strong TRAF1 expression in the tumor cells of most LyP cases (42/49, 84%). In contrast, tumor cells of primary and secondary cALCL revealed TRAF1 expression in only a few cases (3/41, 7%) as shown for sALCL without skin manifestation. The data indicate that TRAF1 expression reliably distinguishes LyP from primary or secondary cALCL. This might be of crucial diagnostic importance and has a strong impact on the treatment decision for patients with cALCL and LyP.


Subject(s)
Ki-1 Antigen/analysis , Lymphoma, Large B-Cell, Diffuse/chemistry , Lymphoma, T-Cell, Cutaneous/chemistry , Lymphoma, T-Cell/chemistry , Lymphomatoid Papulosis/metabolism , TNF Receptor-Associated Factor 1/analysis , Animals , Humans , Mice , Mice, Inbred BALB C , TNF Receptor-Associated Factor 1/immunology , TNF Receptor-Associated Factor 1/physiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...