Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cells ; 10(8)2021 08 12.
Article in English | MEDLINE | ID: mdl-34440839

ABSTRACT

Uncontrolled inflammation is associated with neurodegenerative conditions in central nervous system tissues, including the retina and brain. We previously found that the neural retina (NR) plays an important role in retinal immunity. Tumor necrosis factor Receptor-Associated Factor 3 (TRAF3) is a known immune regulator expressed in the retina; however, whether TRAF3 regulates retinal immunity is unknown. We have generated the first conditional NR-Traf3 knockout mouse model (Chx10-Cre/Traf3f/f) to enable studies of neuronal TRAF3 function. Here, we evaluated NR-Traf3 depletion effects on whole retinal TRAF3 protein expression, visual acuity, and retinal structure and function. Additionally, to determine if NR-Traf3 plays a role in retinal immune regulation, we used flow cytometry to assess immune cell infiltration following acute local lipopolysaccharide (LPS) administration. Our results show that TRAF3 protein is highly expressed in the NR and establish that NR-Traf3 depletion does not affect basal retinal structure or function. Importantly, NR-Traf3 promoted LPS-stimulated retinal immune infiltration. Thus, our findings propose NR-Traf3 as a positive regulator of retinal immunity. Further, the NR-Traf3 mouse provides a tool for investigations of neuronal TRAF3 as a novel potential target for therapeutic interventions aimed at suppressing retinal inflammatory disease and may also inform treatment approaches for inflammatory neurodegenerative brain conditions.


Subject(s)
Homeodomain Proteins/genetics , Neurons/metabolism , Retina/metabolism , TNF Receptor-Associated Factor 3/genetics , Transcription Factors/genetics , Animals , Disease Models, Animal , Electroretinography , Immunity/drug effects , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Neurons/immunology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Retina/physiology , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/metabolism , Transcription Factors/deficiency , Uveitis/etiology , Uveitis/immunology , Uveitis/metabolism , Visual Acuity
2.
J Immunol ; 207(1): 322-332, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34145060

ABSTRACT

The adaptor protein TNFR-associated factor 3 (TRAF3) is required for in vivo T cell effector functions and for normal TCR/CD28 signaling. TRAF3-mediated enhancement of TCR function requires engagement of both CD3 and CD28, but the molecular mechanisms underlying how TRAF3 interacts with and impacts TCR/CD28-mediated complexes to enhance their signaling remains an important knowledge gap. We investigated how TRAF3 is recruited to, and regulates, CD28 as a TCR costimulator. Direct association with known signaling motifs in CD28 was dispensable for TRAF3 recruitment; rather, TRAF3 associated with the CD28-interacting protein linker of activated T cells (LAT) in human and mouse T cells. TRAF3-LAT association required the TRAF3 TRAF-C domain and a newly identified TRAF2/3 binding motif in LAT. TRAF3 inhibited function of the LAT-associated negative regulatory protein Dok1, which is phosphorylated at an inhibitory tyrosine residue by the tyrosine kinase breast tumor kinase (Brk/PTK6). TRAF3 regulated Brk activation in T cells, limiting the association of protein tyrosine phosphatase 1B (PTP1B) with the LAT complex. In TRAF3-deficient cells, LAT complex-associated PTP1B was associated with dephosphorylation of Brk at an activating tyrosine residue, potentially reducing its ability to inhibit Dok1. Consistent with these findings, inhibiting PTP1B activity in TRAF3-deficient T cells rescued basal and TCR/CD28-mediated activation of Src family kinases. These results reveal a new mechanism for promotion of TCR/CD28-mediated signaling through restraint of negative regulation of LAT by TRAF3, enhancing the understanding of regulation of the TCR complex.


Subject(s)
CD28 Antigens/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 3/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/genetics
3.
FASEB J ; 34(5): 7144-7159, 2020 05.
Article in English | MEDLINE | ID: mdl-32275117

ABSTRACT

Disrupted mitochondrial function and reactive oxygen species (ROS) generation cause cellular damage and oxidative stress-induced macrophage inflammatory cell death. It remains unclear how mitochondrial dysfunction relates to inflammasome activation and pyroptotic cell death. In this study, we demonstrated that tumor necrosis factor receptor-associated factor 3 (TRAF3) regulates mitochondrial ROS production and promotes TLR agonist LPS plus nigericin (LPS/Ng)-induced inflammasome and pyroptosis in mouse primary macrophages and human monocyte THP-1 cells. Co-IP assays confirmed that TRAF3 forms a complex with TRAF2 and cIAP1 and mediates ubiquitin and degradation of Unc-51 like autophagy activating kinase 1 (ULK1). Moreover, knockdown of ULK1 in THP-1 cells significantly promoted LPS/Ng-induced inflammasome by activating caspase 1 and mature IL-1ß. Apoptosis inducing factor (AIF) translocation from mitochondrial to nuclear was observed in ULK1-deficient THP-1 cells under LPS/Ng stimulation, which mediates LPS/Ng-induced cell death in ULK1 deficient macrophages. In conclusion, this study identified a novel role of TRAF3 in regulation of ULK1 ubiquitination and inflammasome signaling and provided molecular mechanisms by which ubiquitination of ULK1 controls mitochondrial ROS production, inflammasome activity, and AIF-dependent pyroptosis.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , TNF Receptor-Associated Factor 3/metabolism , Animals , Autophagy-Related Protein-1 Homolog/deficiency , Autophagy-Related Protein-1 Homolog/genetics , Cells, Cultured , Gene Knockdown Techniques , Humans , Inflammasomes/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/pharmacology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Monocytes/cytology , Monocytes/metabolism , Nigericin/pharmacology , Pyroptosis/drug effects , Reactive Oxygen Species/metabolism , THP-1 Cells , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/genetics , Ubiquitination/drug effects
4.
Sci Rep ; 9(1): 12884, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31501481

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins/metabolism , TNF Receptor-Associated Factor 3/metabolism , Animals , B-Lymphocytes/metabolism , Cell Line, Tumor , Cell Survival , Humans , Mice , Phosphorylation , STAT3 Transcription Factor/metabolism , TNF Receptor-Associated Factor 3/deficiency
5.
Sci Rep ; 6: 35349, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27752131

ABSTRACT

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.


Subject(s)
B-Lymphocytes/metabolism , Lymphoma/genetics , Multiple Myeloma/genetics , TNF Receptor-Associated Factor 3/genetics , Animals , Cellular Reprogramming/genetics , Glucose/genetics , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Hexokinase/genetics , Hexokinase/metabolism , Lymphoma/pathology , Mice, Knockout , Multiple Myeloma/pathology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/metabolism , NF-kappaB-Inducing Kinase
6.
J Leukoc Biol ; 100(5): 919-926, 2016 11.
Article in English | MEDLINE | ID: mdl-27154354

ABSTRACT

This review summarizes the current state of knowledge regarding the roles of the signaling adapter protein tumor necrosis factor receptor (TNFR)-associated factor 3 in regulating the functions of B and T lymphocytes. In B lymphocytes, TNFR-associated factor 3 inhibits signaling by TNFR superfamily receptors, Toll-like receptors, and interleukin-6R. In contrast, signaling to B cells by the virally encoded oncogenic protein latent membrane protein 1 is promoted by TNFR-associated factor 3. An important B cell-specific role for TNFR-associated factor 3 is the inhibition of homeostatic survival, directly relevant to the common occurrence of TNFR-associated factor 3 mutations in human B cell malignancies. TNFR-associated factor 3 was recently found to be a resident nuclear protein in B cells, where it interacts with and inhibits gene expression mediated by the cAMP response element-binding protein transcription complex, including expression of the prosurvival protein myeloid leukemia cell differentiation protein 1. In T lymphocytes, TNFR-associated factor 3 is required for normal signaling by the T cell antigen receptor, while inhibiting signaling by the interleukin-2 receptor. Cytoplasmic TNFR -associated factor 3 restrains nuclear factor-κB2 activation in both T and B cells. Clinical implications and future directions for the study of this context-dependent signaling regulator are discussed.


Subject(s)
B-Lymphocytes/immunology , Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 3/immunology , Animals , Antibody Formation , CD40 Antigens/immunology , Cell Line , Cell Survival , Forecasting , Humans , Immunity, Innate , Mice , Mice, Knockout , NF-kappa B/metabolism , Nuclear Proteins/immunology , Protein Kinase C-delta/immunology , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Receptors, Cytokine/immunology , TNF Receptor-Associated Factor 3/deficiency , Viral Matrix Proteins/immunology
7.
Leuk Res ; 41: 85-95, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26740054

ABSTRACT

Bortezomib, a clinical drug for multiple myeloma (MM) and mantle cell lymphoma, exhibits complex mechanisms of action, which vary depending on the cancer type and the critical genetic alterations of each cancer. Here we investigated the signaling mechanisms of bortezomib in mouse B lymphoma and human MM cells deficient in a new tumor suppressor gene, TRAF3. We found that bortezomib consistently induced up-regulation of the cell cycle inhibitor p21(WAF1) and the pro-apoptotic protein Noxa as well as cleavage of the anti-apoptotic protein Mcl-1. Interestingly, bortezomib induced the activation of NF-κB1 and the accumulation of the oncoprotein c-Myc, but inhibited the activation of NF-κB2. Furthermore, we demonstrated that oridonin (an inhibitor of NF-κB1 and NF-κB2) or AD 198 (a drug targeting c-Myc) drastically potentiated the anti-cancer effects of bortezomib in TRAF3-deficient malignant B cells. Taken together, our findings increase the understanding of the mechanisms of action of bortezomib, which would aid the design of novel bortezomib-based combination therapies. Our results also provide a rationale for clinical evaluation of the combinations of bortezomib and oridonin (or other inhibitors of NF-κB1/2) or AD 198 (or other drugs targeting c-Myc) in the treatment of lymphoma and MM, especially in patients containing TRAF3 deletions or relevant mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Lymphoma, B-Cell/genetics , Multiple Myeloma/genetics , Signal Transduction/drug effects , TNF Receptor-Associated Factor 3/deficiency , Animals , Disease Models, Animal , Humans , Immunoblotting , Immunoprecipitation , Mice , Proteasome Inhibitors/pharmacology , TNF Receptor-Associated Factor 3/genetics , Transduction, Genetic
8.
Hypertension ; 66(2): 356-67, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26034202

ABSTRACT

Cardiac hypertrophy, a common early symptom of heart failure, is regulated by numerous signaling pathways. Here, we identified tumor necrosis factor receptor-associated factor 3 (TRAF3), an adaptor protein in tumor necrosis factor-related signaling cascades, as a key regulator of cardiac hypertrophy in response to pressure overload. TRAF3 expression was upregulated in hypertrophied mice hearts and failing human hearts. Four weeks after aortic banding, cardiac-specific conditional TRAF3-knockout mice exhibited significantly reduced cardiac hypertrophy, fibrosis, and dysfunction. Conversely, transgenic mice overexpressing TRAF3 in the heart developed exaggerated cardiac hypertrophy in response to pressure overload. TRAF3 also promoted an angiotensin II- or phenylephrine-induced hypertrophic response in isolated cardiomyocytes. Mechanistically, TRAF3 directly bound to TANK-binding kinase 1 (TBK1), causing increased TBK1 phosphorylation in response to hypertrophic stimuli. This interaction between TRAF3 and TBK1 further activated AKT signaling, which ultimately promoted the development of cardiac hypertrophy. Our findings not only reveal a key role of TRAF3 in regulating the hypertrophic response but also uncover TRAF3-TBK1-AKT as a novel signaling pathway in the development of cardiac hypertrophy and heart failure. This pathway may represent a potential therapeutic target for this pathological process.


Subject(s)
Cardiomegaly/pathology , Cardiomegaly/physiopathology , Signal Transduction/physiology , TNF Receptor-Associated Factor 3/physiology , Up-Regulation/physiology , Angiotensin II/pharmacology , Animals , Disease Models, Animal , Female , Humans , Hypertrophy , Male , Mice , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phenylephrine/pharmacology , Phosphorylation/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/genetics
9.
Oncotarget ; 6(24): 20621-35, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26036313

ABSTRACT

Despite the introduction of new treatment options for multiple myeloma (MM), a majority of patients relapse due to the development of resistance. Unraveling new mechanisms underlying resistance could lead to identification of possible targets for combinatorial treatment. Using TRAF3 deleted/mutated MM cell lines, we evaluated the role of the cellular inhibitor of apoptosis 2 (cIAP2) in drug resistance and uncovered the plausible mechanisms underlying this resistance and possible strategies to overcome this by combinatorial treatment. In MM, cIAP2 is part of the gene signature of aberrant NF-κB signaling and is heterogeneously expressed amongst MM patients. In cIAP2 overexpressing cells a decreased sensitivity to the proteasome inhibitors bortezomib, MG132 and carfilzomib was observed. Gene expression analysis revealed that 440 genes were differentially expressed due to cIAP2 overexpression. Importantly, the data imply that cIAPs are rational targets for combinatorial treatment in the population of MM with deleted/mutated TRAF3. Indeed, we found that treatment with the IAP inhibitor AT-406 enhanced the anti-MM effect of bortezomib in the investigated cell lines. Taken together, our results show that cIAP2 is an important factor mediating bortezomib resistance in MM cells harboring TRAF3 deletion/mutation and therefore should be considered as a target for combinatorial treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Proteasome Inhibitors/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Azocines/administration & dosage , Azocines/pharmacology , Baculoviral IAP Repeat-Containing 3 Protein , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/pharmacology , Bortezomib/administration & dosage , Bortezomib/pharmacology , Case-Control Studies , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , HEK293 Cells , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , Multiple Myeloma/pathology , NF-kappa B/metabolism , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
10.
Cell Cycle ; 14(8): 1156-63, 2015.
Article in English | MEDLINE | ID: mdl-25723057

ABSTRACT

Tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) is broadly involved in different receptor-mediated signaling pathways. Considerable progress was made recently in understanding the role of TRAF3 in T cell biology. Here we review these new findings about how TRAF3 participates in T cell development and function. The different roles of TRAF3 in distinct immune cells are also compared. That TRAF3 is required for T cell effector functions, and invariant Natural Killer T cell function and development, was unexpected. Another surprising finding is that TRAF3 normally restrains regulatory T cell development. It is now clear that TRAF3 regulates signaling to T cells not only through costimulatory members of the TNFR superfamily, but also through the T cell receptor complex, and cytokine receptors. The diverse roles it plays support the multifaceted nature of this molecule. How TRAF3 mediates integration of different signaling cascades is an important topic for future study.


Subject(s)
T-Lymphocytes/metabolism , TNF Receptor-Associated Factor 3/metabolism , Animals , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NF-kappa B/metabolism , Receptors, Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/genetics
11.
J Hematol Oncol ; 7: 56, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25200342

ABSTRACT

BACKGROUND: Identification of novel genetic risk factors is imperative for a better understanding of B lymphomagenesis and for the development of novel therapeutic strategies. TRAF3, a critical regulator of B cell survival, was recently recognized as a tumor suppressor gene in B lymphocytes. The present study aimed to identify novel oncogenes involved in malignant transformation of TRAF3-deficient B cells. METHODS: We used microarray analysis to identify genes differentially expressed in TRAF3-/- mouse splenic B lymphomas. We employed lentiviral vector-mediated knockdown or overexpression to manipulate gene expression in human multiple myeloma (MM) cell lines. We analyzed cell apoptosis and proliferation using flow cytometry, and performed biochemical studies to investigate signaling mechanisms. To delineate protein-protein interactions, we applied affinity purification followed by mass spectrometry-based sequencing. RESULTS: We identified mutated in colorectal cancer (MCC) as a gene strikingly up-regulated in TRAF3-deficient mouse B lymphomas and human MM cell lines. Aberrant up-regulation of MCC also occurs in a variety of primary human B cell malignancies, including non-Hodgkin lymphoma (NHL) and MM. In contrast, MCC expression was not detected in normal or premalignant TRAF3-/- B cells even after treatment with B cell stimuli, suggesting that aberrant up-regulation of MCC is specifically associated with malignant transformation of B cells. In elucidating the functional roles of MCC in malignant B cells, we found that lentiviral shRNA vector-mediated knockdown of MCC induced apoptosis and inhibited proliferation in human MM cells. Experiments of knockdown and overexpression of MCC allowed us to identify several downstream targets of MCC in human MM cells, including phospho-ERK, c-Myc, p27, cyclin B1, Mcl-1, caspases 8 and 3. Furthermore, we identified 365 proteins (including 326 novel MCC-interactors) in the MCC interactome, among which PARP1 and PHB2 were two hubs of MCC signaling pathways in human MM cells. CONCLUSIONS: Our results indicate that in sharp contrast to its tumor suppressive role in colorectal cancer, MCC functions as an oncogene in B cells. Our findings suggest that MCC may serve as a diagnostic marker and therapeutic target in B cell malignancies, including NHL and MM.


Subject(s)
B-Lymphocytes/pathology , Cell Transformation, Neoplastic/genetics , Genes, MCC/genetics , Oncogenes/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Chromatin Immunoprecipitation , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Lymphoma, Non-Hodgkin/genetics , Mice , Mice, Knockout , Multiple Myeloma/genetics , Oligonucleotide Array Sequence Analysis , Prohibitins , Reverse Transcriptase Polymerase Chain Reaction , TNF Receptor-Associated Factor 3/deficiency , Tandem Mass Spectrometry
12.
PLoS One ; 9(7): e102120, 2014.
Article in English | MEDLINE | ID: mdl-25010048

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Homeostasis/immunology , Immunologic Memory , TNF Receptor-Associated Factor 3/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Female , Interleukin-15/metabolism , Lymphocyte Activation/immunology , Male , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , TNF Receptor-Associated Factor 3/deficiency
13.
Leuk Res ; 38(3): 393-401, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24418753

ABSTRACT

Using a mouse model with the tumor suppressor TRAF3 deleted from B cells, we identified Sox5 as a gene strikingly up-regulated in B lymphomas. Sox5 proteins were not detected in normal or premalignant TRAF3(-/-) B cells even after treatment with B cell stimuli. The Sox5 expressed in TRAF3(-/-) B lymphomas represents a novel isoform of Sox5, and was localized in the nucleus of malignant B cells. Overexpression of Sox5 inhibited cell cycle progression, and up-regulated the protein levels of p27 and ß-catenin in human multiple myeloma cells. Together, our findings indicate that Sox5 regulates the proliferation of malignant B cells.


Subject(s)
B-Lymphocytes/metabolism , Cell Nucleus/genetics , Gene Expression Regulation, Neoplastic , SOXD Transcription Factors/genetics , TNF Receptor-Associated Factor 3/genetics , Animals , Antibodies/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Cell Cycle/genetics , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Proliferation , Humans , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , SOXD Transcription Factors/metabolism , Signal Transduction , TNF Receptor-Associated Factor 3/deficiency , beta Catenin/genetics , beta Catenin/metabolism
14.
Proc Natl Acad Sci U S A ; 110(52): 21107-12, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24324158

ABSTRACT

Induction of self-tolerance in developing T cells depends on medullary thymic epithelial cells (mTECs), whose development, in turn, requires signals from single-positive (SP) thymocytes. Thus, the absence of SP thymocytes in Tcra(-/-) mice results in a profound deficiency in mTECs. Here, we have probed the mechanism that underlies this requirement for cross-talk with thymocytes in medullary development. Previous studies have implicated nonclassical NF-κB as a pathway important in the development of mTECs, because mice lacking RelB, NIK, or IKKα, critical components of this pathway, have an almost complete absence of mTECs, with resulting autoimmune pathology. We therefore assessed the effect of selective deletion in TEC of TNF receptor-associated factor 3 (TRAF3), an inhibitor of nonclassical NF-κB signaling. Deletion of TRAF3 in thymic epithelial cells allowed RelB-dependent development of normal numbers of AIRE-expressing mTECs in the complete absence of SP thymocytes. Thus, mTEC development can occur in the absence of cross-talk with SP thymocytes, and signals provided by SP T cells are needed to overcome TRAF3-imposed arrest in mTEC development mediated by inhibition of nonclassical NF-κB. We further observed that TRAF3 deletion is also capable of overcoming all requirements for LTßR and CD40, which are otherwise necessary for mTEC development, but is not sufficient to overcome the requirement for RANKL, indicating a role for RANKL that is distinct from the signals provided by SP thymocytes. We conclude that TRAF3 plays a central role in regulation of mTEC development by imposing requirements for SP T cells and costimulation-mediated cross-talk in generation of the medullary compartment.


Subject(s)
Cell Differentiation/immunology , Receptor Cross-Talk/immunology , Self Tolerance/immunology , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 3/immunology , Thymocytes/metabolism , Animals , CD40 Antigens/genetics , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , TNF Receptor-Associated Factor 3/deficiency , Thymocytes/immunology
15.
BMC Cancer ; 13: 481, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24131623

ABSTRACT

BACKGROUND: TRAF3, a new tumor suppressor identified in human non-Hodgkin lymphoma (NHL) and multiple myeloma (MM), induces PKCδ nuclear translocation in B cells. The present study aimed to evaluate the therapeutic potential of two PKCδ activators, N-Benzyladriamycin-14-valerate (AD 198) and ingenol-3-angelate (PEP005), on NHL and MM. METHODS: In vitro anti-tumor activities of AD 198 and PEP005 were determined using TRAF3-/- mouse B lymphoma and human patient-derived MM cell lines as model systems. In vivo therapeutic effects of AD 198 were assessed using NOD SCID mice transplanted with TRAF3-/- mouse B lymphoma cells. Biochemical studies were performed to investigate signaling mechanisms induced by AD 198 or PEP005, including subcellular translocation of PKCδ. RESULTS: We found that AD 198 exhibited potent in vitro and in vivo anti-tumor activity on TRAF3-/- tumor B cells, while PEP005 displayed contradictory anti- or pro-tumor activities on different cell lines. Detailed mechanistic investigation revealed that AD 198 did not affect PKCδ nuclear translocation, but strikingly suppressed c-Myc expression and inhibited the phosphorylation of ERK, p38 and JNK in TRAF3-/- tumor B cells. In contrast, PEP005 activated multiple signaling pathways in these cells, including PKCδ, PKCα, PKCε, NF-κB1, ERK, JNK, and Akt. Additionally, AD198 also potently inhibited the proliferation/survival and suppressed c-Myc expression in TRAF3-sufficient mouse and human B lymphoma cell lines. Furthermore, we found that reconstitution of c-Myc expression conferred partial resistance to the anti-proliferative/apoptosis-inducing effects of AD198 in human MM cells. CONCLUSIONS: AD 198 and PEP005 have differential effects on malignant B cells through distinct biochemical mechanisms. Our findings uncovered a novel, PKCδ-independent mechanism of the anti-tumor effects of AD 198, and suggest that AD 198 has therapeutic potential for the treatment of NHL and MM involving TRAF3 inactivation or c-Myc up-regulation.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/analogs & derivatives , Lymphoma, B-Cell/genetics , Multiple Myeloma/genetics , TNF Receptor-Associated Factor 3/deficiency , Animals , Antibiotics, Antineoplastic/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Diterpenes/administration & dosage , Diterpenes/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Genetic Vectors/genetics , Humans , Isografts , Lentivirus/genetics , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/mortality , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Protein Kinase C-delta/metabolism , Protein Transport , Proteolysis/drug effects , Proto-Oncogene Proteins c-myc/genetics , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Transduction, Genetic
16.
J Leukoc Biol ; 90(6): 1149-57, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21971520

ABSTRACT

The key role of TRAF6 in TLR signaling pathways is well known. More recent evidence has implicated TRAF3 as another TRAF family member important to certain TLR responses of myeloid cells. Previous studies demonstrate that TRAF3 functions are highly context-dependent, displaying receptor and cell-type specificity. We thus examined the TLR responses of TRAF3(-/-)mouse B lymphocytes to test the hypothesis that TRAF3 plays distinct roles in such responses, depending on cell type. TRAF3(-/-) DC are known to have a defect in type 1 IFN production and here, showed diminished production of TNF and IL-10 and unaltered IL-6. In marked contrast, TRAF3(-/-) B cells made elevated amounts of TNF and IL-6 protein, as well as IL-10 and IP-10 mRNA, in response to TLR ligands. Also, in contrast to TRAF3(-/-) DC, the type 1 IFN pathway was elevated in TRAF3(-/-) B cells. Increased early responses of TRAF3(-/-) B cells to TLR signals were independent of cell survival or proliferation but associated with elevated canonical NF-κB activation. Additionally, TRAF3(-/-) B cells displayed enhanced TLR-mediated expression of AID and Ig isotype switching. Thus, TRAF3 plays varied and cell type-specific, biological roles in TLR responses.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/physiology , Toll-Like Receptors/biosynthesis , Animals , B-Lymphocyte Subsets/cytology , Cells, Cultured , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology
17.
J Immunol ; 186(1): 143-55, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21084666

ABSTRACT

We recently reported that TNFR-associated factor (TRAF)3, a ubiquitously expressed adaptor protein, promotes mature B cell apoptosis. However, the specific function of TRAF3 in T cells has remained unclear. In this article, we report the generation and characterization of T cell-specific TRAF3(-/-) mice, in which the traf3 gene was deleted from thymocytes and T cells. Ablation of TRAF3 in the T cell lineage did not affect CD4 or CD8 T cell populations in secondary lymphoid organs or the numbers or proportions of CD4(+),CD8(+) or double-positive or double-negative thymocytes, except that the T cell-specific TRAF3(-/-) mice had a 2-fold increase in FoxP3(+) T cells. In striking contrast to mice lacking TRAF3 in B cells, the T cell TRAF3-deficient mice exhibited defective IgG1 responses to a T-dependent Ag, as well as impaired T cell-mediated immunity to infection with Listeria monocytogenes. Surprisingly, we found that TRAF3 was recruited to the TCR/CD28 signaling complex upon costimulation and that TCR/CD28-mediated proximal and distal signaling events were compromised by TRAF3 deficiency. These findings provide insights into the roles played by TRAF3 in T cell activation and T cell-mediated immunity.


Subject(s)
CD28 Antigens/physiology , Immunity, Cellular , Receptors, Antigen, T-Cell/physiology , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , TNF Receptor-Associated Factor 3/physiology , Animals , Antibodies, Bacterial/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cells, Cultured , Immunity, Cellular/genetics , Immunoglobulin G/biosynthesis , Listeriosis/genetics , Listeriosis/immunology , Listeriosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/genetics , T-Lymphocytes/pathology , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/genetics
18.
J Immunol ; 185(11): 6555-62, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21041727

ABSTRACT

CD40 is required for T cell-dependent humoral immunity, but it can also contribute to the pathogenesis of autoimmunity and B cell malignancy. The TNFR-associated factor (TRAF)2 and TRAF6 adaptor proteins are positive regulators of CD40 signaling required to activate downstream kinase cascades and transcription factors. In contrast, TRAF3 can serve as a negative regulator of CD40 signaling, and CD40 signals are amplified in TRAF3(-/-) B cells. We previously reported a gain-of-function polymorphism of the human CD40 receptor, hCD40-P227A, which signals in an amplified manner to B lymphocytes. In this study, we show that hCD40-P227A binds more TRAF3 and TRAF5, as well as certain associated proteins, than wild-type-CD40. Studies in TRAF-deficient B cell lines revealed that hCD40-P227A uses TRAF3 as a positive rather than negative regulator. Although located outside of any known TRAF binding sites, the P227A polymorphism can alter TRAF binding and dramatically changes the role played by TRAF3 in CD40 signaling.


Subject(s)
CD40 Antigens/antagonists & inhibitors , CD40 Antigens/metabolism , Genetic Variation/immunology , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/metabolism , Up-Regulation/immunology , Amino Acid Sequence , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , Binding Sites/genetics , Binding Sites/immunology , CD40 Antigens/genetics , Cell Line , Clone Cells , Humans , Interleukin-6/biosynthesis , Mice , Molecular Sequence Data , Polymorphism, Genetic/immunology , Signal Transduction/genetics , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/physiology , TNF Receptor-Associated Factor 6/deficiency , TNF Receptor-Associated Factor 6/genetics , Up-Regulation/genetics
19.
J Virol ; 84(13): 6605-14, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20410275

ABSTRACT

Epstein-Barr Virus (EBV) latent membrane protein 1 (LMP1) is required for EBV B-lymphocyte transformation, transforms rodent fibroblasts, and can induce lymphoma and epithelial hyperplasia in transgenic mice. Two domains have been identified within the intracellular carboxy terminus that can activate NF-kappaB, C-terminus-activating region 1 (CTAR1) and CTAR2, through interactions with tumor necrosis receptor-associated factors (TRAFs). CTAR1 can activate both the canonical and noncanonical NF-kappaB pathways and has unique effects on cellular gene expression. The epidermal growth factor receptor (EGFR) is highly induced by LMP1-CTAR1 in epithelial cells through activation of a novel NF-kappaB form containing p50 homodimers and Bcl-3. To further understand the regulation of NF-kappaB in CTAR1-induced EGFR expression, we evaluated the ability of CTAR1 to induce EGFR in mouse embryonic fibroblasts (MEFs) defective for different NF-kappaB effectors. CTAR1-mediated EGFR induction required the NF-kappaB-inducing kinase (NIK) but not the IkappaB kinase (IKK) complex components that regulate canonical or noncanonical NF-kappaB pathways. CTAR1-mediated induction of nuclear p50 occurred in IKKbeta-, IKKgamma-, and NIK-defective MEFs, indicating that this induction is not dependent on the canonical or noncanonical NF-kappaB pathways. EGFR and nuclear p50 were expressed at high levels in TRAF2(-/-) fibroblasts and were not induced by CTAR1. In TRAF3(-/-) MEFs, CTAR1 induced nuclear p50 but did not affect basal levels of STAT3 serine phosphorylation or induce EGFR expression. EGFR was induced by LMP1 in TRAF6(-/-) MEFs. These findings suggest that this novel NF-kappaB pathway is differentially regulated by TRAF2 and TRAF3, and that distinct interactions of LMP1 and its effectors regulate LMP1-mediated gene expression.


Subject(s)
ErbB Receptors/biosynthesis , Herpesvirus 4, Human/physiology , NF-kappa B p50 Subunit/metabolism , Viral Matrix Proteins/metabolism , Virus Attachment , Animals , Fibroblasts/virology , I-kappa B Kinase/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/metabolism , STAT3 Transcription Factor/metabolism , TNF Receptor-Associated Factor 2/deficiency , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/metabolism , TNF Receptor-Associated Factor 6/deficiency , TNF Receptor-Associated Factor 6/metabolism , NF-kappaB-Inducing Kinase
20.
Immunol Res ; 39(1-3): 22-32, 2007.
Article in English | MEDLINE | ID: mdl-17917053

ABSTRACT

Members of the tumor necrosis factor receptor (TNFR) superfamily employ cytoplasmic adapter proteins called TNF-R associated factors (TRAF) to initiate and regulate signaling pathways. Although many of these receptors associate with TRAF3, it has been unclear how this TRAF functions in immune responses. New information appearing through the use of novel experimental models reveals that TRAF3 can mediate both activating and inhibitory signals, and can participate in regulation of multiple members of the TNFR superfamily. TRAF3 is also important for signaling via innate immune receptors, as well as an oncogenic mimic of a normal receptor that is implicated in promoting both malignancies and autoimmunity.


Subject(s)
CD40 Antigens/metabolism , Lymphocytes/immunology , Receptors, Tumor Necrosis Factor/metabolism , TNF Receptor-Associated Factor 3/metabolism , Animals , CD40 Antigens/immunology , Humans , Lymphocytes/metabolism , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...