1
|
Zhang L, Dou X, Zheng Z, Ye C, Lu TX, Liang HL, Wang L, Weichselbaum RR, He C. YTHDF2/m 6 A/NF-κB axis controls anti-tumor immunity by regulating intratumoral Tregs. EMBO J 2023; 42:e113126. [PMID: 37345898 PMCID: PMC10390869 DOI: 10.15252/embj.2022113126] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023] Open
Abstract
N6 -methyladenosine (m6 A) in messenger RNA (mRNA) regulates immune cells in homeostasis and in response to infection and inflammation. The function of the m6 A reader YTHDF2 in the tumor microenvironment (TME) in these contexts has not been explored. We discovered that the loss of YTHDF2 in regulatory T (Treg) cells reduces tumor growth in mice. Deletion of Ythdf2 in Tregs does not affect peripheral immune homeostasis but leads to increased apoptosis and impaired suppressive function of Treg cells in the TME. Elevated tumor necrosis factor (TNF) signaling in the TME promotes YTHDF2 expression, which in turn regulates NF-κB signaling by accelerating the degradation of m6 A-modified transcripts that encode NF-κB-negative regulators. This TME-specific regulation of Treg by YTHDF2 points to YTHDF2 as a potential target for anti-cancer immunotherapy, where intratumoral Treg cells can be targeted to enhance anti-tumor immune response while avoiding Treg cells in the periphery to minimize undesired inflammations.
Collapse
Affiliation(s)
- Linda Zhang
- Department of ChemistryThe University of ChicagoChicagoILUSA
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoILUSA
- Institute for Biophysical DynamicsThe University of ChicagoChicagoILUSA
- Howard Hughes Medical InstituteUniversity of ChicagoChicagoILUSA
| | - Xiaoyang Dou
- Department of ChemistryThe University of ChicagoChicagoILUSA
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoILUSA
- Institute for Biophysical DynamicsThe University of ChicagoChicagoILUSA
- Howard Hughes Medical InstituteUniversity of ChicagoChicagoILUSA
| | - Zhong Zheng
- Department of ChemistryThe University of ChicagoChicagoILUSA
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoILUSA
- Institute for Biophysical DynamicsThe University of ChicagoChicagoILUSA
- Howard Hughes Medical InstituteUniversity of ChicagoChicagoILUSA
| | - Chang Ye
- Department of ChemistryThe University of ChicagoChicagoILUSA
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoILUSA
- Institute for Biophysical DynamicsThe University of ChicagoChicagoILUSA
- Howard Hughes Medical InstituteUniversity of ChicagoChicagoILUSA
| | - Thomas X Lu
- Department of ChemistryThe University of ChicagoChicagoILUSA
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoILUSA
- Institute for Biophysical DynamicsThe University of ChicagoChicagoILUSA
- Present address:
Southern Indiana PhysiciansIndiana University HealthBloomingtonINUSA
| | - Hua L Liang
- Department of Radiation and Cellular OncologyUniversity of ChicagoChicagoILUSA
- Ludwig Center for Metastasis ResearchUniversity of ChicagoChicagoILUSA
| | - Liangliang Wang
- Department of Radiation and Cellular OncologyUniversity of ChicagoChicagoILUSA
- Ludwig Center for Metastasis ResearchUniversity of ChicagoChicagoILUSA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular OncologyUniversity of ChicagoChicagoILUSA
- Ludwig Center for Metastasis ResearchUniversity of ChicagoChicagoILUSA
| | - Chuan He
- Department of ChemistryThe University of ChicagoChicagoILUSA
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoILUSA
- Institute for Biophysical DynamicsThe University of ChicagoChicagoILUSA
- Howard Hughes Medical InstituteUniversity of ChicagoChicagoILUSA
| |
Collapse
|
2
|
Hornick EL, Bishop GA. TRAF3: Guardian of T lymphocyte functions. Front Immunol 2023; 14:1129251. [PMID: 36814922 PMCID: PMC9940752 DOI: 10.3389/fimmu.2023.1129251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/27/2023] [Indexed: 02/08/2023] Open
Abstract
Tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) is an adapter protein with many context-specific functions. Early studies of lymphocyte TRAF3 hinted at TRAF3's importance for T cell function, but elucidation of specific mechanisms was delayed by early lethality of globally TRAF3-/- mice. Development of a conditional TRAF3-deficient mouse enabled important descriptive and mechanistic insights into how TRAF3 promotes optimal T cell function. Signaling through the T cell antigen receptor (TCR) fails to induce normal proliferation and survival in TRAF3 -/- T cells, and TCR-activated cells in vitro and in vivo have deficient cytokine production. These defects can be traced to incorrect localization and function of negative regulatory phosphatases acting at different parts of the signaling cascade, as can dysregulated effector responses and memory T cell homeostasis in vivo and an enlarged regulatory T cell (Treg) compartment. The important regulatory activity of TRAF3 is also evident at members of the TNFR superfamily and cytokine receptors. Here, we review significant advances in mechanistic understanding of how TRAF3 regulates T cell differentiation and function, through modulation of signaling through the TCR, costimulatory receptors, and cytokine receptors. Finally, we briefly discuss the recent identification of families carrying single allele loss-of-function mutations in TRAF3, and compare the findings in their T cells with the T cell defects identified in mice whose T cells completely lack T cell TRAF3. Together, the body of work describing TRAF3-mediated regulation of T cell effector function and differentiation frame TRAF3 as an important modulator of T cell signal integration.
Collapse
Affiliation(s)
- Emma L Hornick
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| | - Gail A Bishop
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States.,Department of Internal Medicine, The University of Iowa, Iowa City, IA, United States.,Research, Iowa City Veterans Affairs Medical Center, Iowa City, IA, United States
| |
Collapse
|
3
|
Gissler MC, Stachon P, Wolf D, Marchini T. The Role of Tumor Necrosis Factor Associated Factors (TRAFs) in Vascular Inflammation and Atherosclerosis. Front Cardiovasc Med 2022; 9:826630. [PMID: 35252400 PMCID: PMC8891542 DOI: 10.3389/fcvm.2022.826630] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/27/2022] [Indexed: 12/20/2022] Open
Abstract
TNF receptor associated factors (TRAFs) represent a family of cytoplasmic signaling adaptor proteins that regulate, bundle, and transduce inflammatory signals downstream of TNF- (TNF-Rs), interleukin (IL)-1-, Toll-like- (TLRs), and IL-17 receptors. TRAFs play a pivotal role in regulating cell survival and immune cell function and are fundamental regulators of acute and chronic inflammation. Lately, the inhibition of inflammation by anti-cytokine therapy has emerged as novel treatment strategy in patients with atherosclerosis. Likewise, growing evidence from preclinical experiments proposes TRAFs as potent modulators of inflammation in atherosclerosis and vascular inflammation. Yet, TRAFs show a highly complex interplay between different TRAF-family members with partially opposing and overlapping functions that are determined by the level of cellular expression, concomitant signaling events, and the context of the disease. Therefore, inhibition of specific TRAFs may be beneficial in one condition and harmful in others. Here, we carefully discuss the cellular expression and signaling events of TRAFs and evaluate their role in vascular inflammation and atherosclerosis. We also highlight metabolic effects of TRAFs and discuss the development of TRAF-based therapeutics in the future.
Collapse
Affiliation(s)
- Mark Colin Gissler
- Cardiology and Angiology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Peter Stachon
- Cardiology and Angiology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Dennis Wolf
- Cardiology and Angiology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- *Correspondence: Dennis Wolf
| | - Timoteo Marchini
- Cardiology and Angiology, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| |
Collapse
|
4
|
Konstorum A, Vella AT, Adler AJ, Laubenbacher RC. A mathematical model of combined CD8 T cell costimulation by 4-1BB (CD137) and OX40 (CD134) receptors. Sci Rep 2019; 9:10862. [PMID: 31350431 PMCID: PMC6659676 DOI: 10.1038/s41598-019-47333-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 07/11/2019] [Indexed: 02/07/2023] Open
Abstract
Combined agonist stimulation of the TNFR costimulatory receptors 4-1BB (CD137) and OX40(CD134) has been shown to generate supereffector CD8 T cells that clonally expand to greater levels, survive longer, and produce a greater quantity of cytokines compared to T cells stimulated with an agonist of either costimulatory receptor individually. In order to understand the mechanisms for this effect, we have created a mathematical model for the activation of the CD8 T cell intracellular signaling network by mono- or dual-costimulation. We show that supereffector status is generated via downstream interacting pathways that are activated upon engagement of both receptors, and in silico simulations of the model are supported by published experimental results. The model can thus be used to identify critical molecular targets of T cell dual-costimulation in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Anna Konstorum
- Center for Quantitative Medicine, School of Medicine, UConn Health, 263 Farmington Ave., Farmington, CT, USA.
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave., Farmington, CT, USA
| | - Adam J Adler
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave., Farmington, CT, USA
| | - Reinhard C Laubenbacher
- Center for Quantitative Medicine, School of Medicine, UConn Health, 263 Farmington Ave., Farmington, CT, USA.,Jackson Laboratory for Genomic Medicine, 263 Farmington Ave., Farmington, CT, USA
| |
Collapse
|
5
|
Pedros C, Altman A, Kong KF. Role of TRAFs in Signaling Pathways Controlling T Follicular Helper Cell Differentiation and T Cell-Dependent Antibody Responses. Front Immunol 2018; 9:2412. [PMID: 30405612 PMCID: PMC6204373 DOI: 10.3389/fimmu.2018.02412] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 09/28/2018] [Indexed: 01/02/2023] Open
Abstract
Follicular helper T (TFH) cells represent a highly specialized CD4+ T cell subpopulation that supports the generation of germinal centers (GC) and provides B cells with critical signals promoting antibody class switching, generation of high affinity antibodies, and memory formation. TFH cells are characterized by the expression of the chemokine receptor CXCR5, the transcription factor Bcl-6, costimulatory molecules ICOS, and PD-1, and the production of cytokine IL-21. The acquisition of a TFH phenotype is a complex and multistep process that involves signals received through engagement of the TCR along with a multitude of costimulatory molecules and cytokines receptors. Members of the Tumor necrosis factor Receptor Associated Factors (TRAF) represent one of the major classes of signaling mediators involved in the differentiation and functions of TFH cells. TRAF molecules are the canonical adaptor molecules that physically interact with members of the Tumor Necrosis Factor Receptor Superfamily (TNFRSF) and actively modulate their downstream signaling cascades through their adaptor function and/or E3 ubiquitin ligase activity. OX-40, GITR, and 4-1BB are the TRAF-dependent TNFRSF members that have been implicated in the differentiation and functions of TFH cells. On the other hand, emerging data demonstrate that TRAF proteins also participate in signaling from the TCR and CD28, which deliver critical signals leading to the differentiation of TFH cells. More intriguingly, we recently showed that the cytoplasmic tail of ICOS contains a conserved TANK-binding kinase 1 (TBK1)-binding motif that is shared with TBK1-binding TRAF proteins. The presence of this TRAF-mimicking signaling module downstream of ICOS is required to mediate the maturation step during TFH differentiation. In addition, JAK-STAT pathways emanating from IL-2, IL-6, IL-21, and IL-27 cytokine receptors affect TFH development, and crosstalk between TRAF-mediated pathways and the JAK-STAT pathways can contribute to generate integrated signals required to drive and sustain TFH differentiation. In this review, we will introduce the molecular interactions and the major signaling pathways controlling the differentiation of TFH cells. In each case, we will highlight the contributions of TRAF proteins to these signaling pathways. Finally, we will discuss the role of individual TRAF proteins in the regulation of T cell-dependent humoral responses.
Collapse
Affiliation(s)
- Christophe Pedros
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Kok-Fai Kong
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| |
Collapse
|
6
|
Roles for TNF-receptor associated factor 3 (TRAF3) in lymphocyte functions. Cytokine Growth Factor Rev 2013; 25:147-56. [PMID: 24433987 DOI: 10.1016/j.cytogfr.2013.12.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/15/2013] [Indexed: 12/27/2022]
Abstract
TRAF3 is an adapter protein that serves and regulates the functions of several types of receptors, located both inside the cell and at the plasma membrane. These include members of the TNF receptor superfamily (TNFR-SF), toll-like receptors (TLR), and cytokine receptors. It has become increasingly evident that the roles and functions of TRAF3 are highly context-dependent. TRAF3 can serve distinct roles for different receptors in the same cell, and also has highly cell-type-dependent functions. This review focuses upon the current state of knowledge regarding how TRAF3 regulates the biology and effector functions of B and T lymphocytes, two major cell types of the adaptive immune response in which TRAF3 has markedly distinct roles.
Collapse
|
7
|
Hildebrand JM, Yi Z, Buchta CM, Poovassery J, Stunz LL, Bishop GA. Roles of tumor necrosis factor receptor associated factor 3 (TRAF3) and TRAF5 in immune cell functions. Immunol Rev 2012; 244:55-74. [PMID: 22017431 DOI: 10.1111/j.1600-065x.2011.01055.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A large and diverse group of receptors utilizes the family of cytoplasmic signaling proteins known as tumor necrosis factor receptor (TNFR)-associated factors (TRAFs). In recent years, there has been a resurgence of interest and exploration of the roles played by TRAF3 and TRAF5 in cellular regulation, particularly in cells of the immune system, the cell types of focus in this review. This work has revealed that TRAF3 and TRAF5 can play diverse roles for different receptors even in the same cell type, as well as distinct roles in different cell types. Evidence indicates that TRAF3 and TRAF5 play important roles beyond the TNFR-superfamily (SF) and viral mimics of its members, mediating certain innate immune receptor and cytokine receptor signals, and most recently, signals delivered by the T-cell receptor (TCR) signaling complex. Additionally, much research has demonstrated the importance of TRAF3-mediated cellular regulation via its cytoplasmic interactions with additional signaling proteins. In particular, we discuss below evidence for the participation by TRAF3 in a number of the regulatory post-translational modifications involving ubiquitin that are important in various signaling pathways.
Collapse
Affiliation(s)
- Joanne M Hildebrand
- Department of Microbiology, The University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | |
Collapse
|
8
|
Murray SE, Polesso F, Rowe AM, Basak S, Koguchi Y, Toren KG, Hoffmann A, Parker DC. NF-κB–inducing kinase plays an essential T cell–intrinsic role in graft-versus-host disease and lethal autoimmunity in mice. J Clin Invest 2011; 121:4775-86. [PMID: 22045568 PMCID: PMC3223068 DOI: 10.1172/jci44943] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 09/21/2011] [Indexed: 11/17/2022] Open
Abstract
NF-κB–inducing kinase (NIK) is an essential upstream kinase in noncanonical NF-κB signaling. NIK-dependent NF-κB activation downstream of several TNF receptor family members mediates lymphoid organ development and B cell homeostasis. Peripheral T cell populations are normal in the absence of NIK, but the role of NIK during in vivo T cell responses to antigen has been obscured by other developmental defects in NIK-deficient mice. Here, we have identified a T cell–intrinsic requirement for NIK in graft-versus-host disease (GVHD), wherein NIK-deficient mouse T cells transferred into MHC class II mismatched recipients failed to cause GVHD. Although NIK was not necessary for antigen receptor signaling, it was absolutely required for costimulation through the TNF receptor family member OX40 (also known as CD134). When we conditionally overexpressed NIK in T cells, mice suffered rapid and fatal autoimmunity characterized by hyperactive effector T cells and poorly suppressive Foxp3(+) Tregs. Together, these data illuminate a critical T cell–intrinsic role for NIK during immune responses and suggest that its tight regulation is critical for avoiding autoimmunity.
Collapse
Affiliation(s)
- Susan E Murray
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon 97239, USA.
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Redmond WL, Ruby CE, Weinberg AD. The role of OX40-mediated co-stimulation in T-cell activation and survival. Crit Rev Immunol 2009; 29:187-201. [PMID: 19538134 DOI: 10.1615/critrevimmunol.v29.i3.10] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The extent of T-cell activation, proliferation, and survival that follows T-cell receptor (TCR) ligation is controlled by several factors, including the strength of TCR stimulation, the availability of prosurvival cytokines, and the presence or absence of co-stimulatory signals. In addition to engagement of the CD28 co-stimulatory receptor by its natural ligands, B7.1 (CD80) and B7.2 (CD86), recent work has begun to elucidate the mechanisms by which signaling through the OX40 (CD134) co-stimulatory receptor, a member of the tumor necrosis factor receptor (TNFR) superfamily, affects T-cell responses. Importantly, OX40 ligation has been shown to augment CD4 and CD8 T-cell clonal expansion, effector differentiation, survival, and in some cases, abrogate the suppressive activity of regulatory FoxP3+CD25+CD4+ T cells. In this review, we focus on the mechanisms regulating OX40 expression on activated T cells as well as the role of OX40-mediated co-stimulation in boosting T-cell clonal expansion, effector differentiation, and survival.
Collapse
Affiliation(s)
- William L Redmond
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Research Center, Providence Portland Medical Center, 4805 NE Glisan St., No. 5F37, Portland, OR 97213, USA
| | | | | |
Collapse
|
10
|
Croft M, So T, Duan W, Soroosh P. The significance of OX40 and OX40L to T-cell biology and immune disease. Immunol Rev 2009; 229:173-91. [PMID: 19426222 DOI: 10.1111/j.1600-065x.2009.00766.x] [Citation(s) in RCA: 443] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SUMMARY OX40 (CD134) and its binding partner, OX40L (CD252), are members of the tumor necrosis factor receptor/tumor necrosis factor superfamily and are expressed on activated CD4(+) and CD8(+) T cells as well as on a number of other lymphoid and non-lymphoid cells. Costimulatory signals from OX40 to a conventional T cell promote division and survival, augmenting the clonal expansion of effector and memory populations as they are being generated to antigen. OX40 additionally suppresses the differentiation and activity of T-regulatory cells, further amplifying this process. OX40 and OX40L also regulate cytokine production from T cells, antigen-presenting cells, natural killer cells, and natural killer T cells, and modulate cytokine receptor signaling. In line with these important modulatory functions, OX40-OX40L interactions have been found to play a central role in the development of multiple inflammatory and autoimmune diseases, making them attractive candidates for intervention in the clinic. Conversely, stimulating OX40 has shown it to be a candidate for therapeutic immunization strategies for cancer and infectious disease. This review provides a broad overview of the biology of OX40 including the intracellular signals from OX40 that impact many aspects of immune function and have promoted OX40 as one of the most prominent costimulatory molecules known to control T cells.
Collapse
Affiliation(s)
- Michael Croft
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
11
|
Growth and Differentiation Advantages of CD4+OX40+ T Cells from Allogeneic Hematopoietic Stem Cell Transplantation Recipients. Biol Blood Marrow Transplant 2008; 14:268-81. [DOI: 10.1016/j.bbmt.2007.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Accepted: 12/05/2007] [Indexed: 11/19/2022]
|
12
|
Rimondi E, Zweyer M, Ricci E, Fadda R, Secchiero P. Receptor activator of nuclear factor kappa B ligand (RANKL) modulates the expression of genes involved in apoptosis and cell cycle in human osteoclasts. Anat Rec (Hoboken) 2007; 290:838-45. [PMID: 17506059 DOI: 10.1002/ar.20550] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It has been clearly established that receptor activator of nuclear factor kappa B ligand (RANKL) is a key cytokine involved in the differentiation of osteoclastic precursors of the monocytic/macrophagic lineage. However, relatively little information is available on the ability of RANKL to modulate the expression of genes controlling cell survival/apoptosis and proliferation in human osteoclastic cells in comparison to macrophages. For this purpose, CD14+ human peripheral blood mononuclear cells, which express the cognate high affinity receptor activator of nuclear factor kappa B (RANK), were differentiated along the macrophagic or osteoclastic lineage by adding macrophage-colony stimulating factor (M-CSF) or M-CSF plus RANKL in culture for 12 days. RANKL up-regulated the expression of the chemokine MIP1alpha, which potentiates osteoclastic differentiation and simultaneously activated both anti-apoptotic (Bcl-2) and pro-apoptotic (CIDEB, PYCARD, and BAK-1) genes. Moreover, RANKL markedly up-regulated cylin D2, while it significantly decreased the levels of cyclin A, cyclin-dependent kinase 2, and other cyclin-dependent kinases, in keeping with the notion that end-stage osteoclasts are nondividing cells. Finally, a long-term exposure of RANKL up-regulated the adaptor protein TRAF3 but not TRAF6.
Collapse
Affiliation(s)
- Erika Rimondi
- Department of Morphology and Embryology, Human Anatomy Section,University of Ferrara, Ferrara, Italy
| | | | | | | | | |
Collapse
|
13
|
Lu LF, Ahonen CL, Lind EF, Raman VS, Cook WJ, Lin LL, Noelle RJ. The in vivo function of a noncanonical TRAF2-binding domain in the C-terminus of CD40 in driving B-cell growth and differentiation. Blood 2007; 110:193-200. [PMID: 17360936 PMCID: PMC1896113 DOI: 10.1182/blood-2006-07-038414] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The recruitment of tumor necrosis factor receptor-associated factors (TRAFs) 1, 2, 3, 5, and 6 to the CD40 cytoplasmic tail upon CD40 trimerization results in downstream signaling events that ultimately lead to CD40-dependent, thymus-dependent (TD) humoral immune responses. Previously, we have shown signaling through the C-terminal tail of CD40 in the absence of canonical TRAF-binding sites is capable of signaling through an alternative TRAF2-binding site. Here, we demonstrate that B cells from mice harboring CD40 with only the C-terminal tail can activate both canonical and noncanonical NFkappaB signaling pathways. Moreover, while lacking germinal center formation, several hallmarks of humoral immune responses including clonal B-cell activation/expansion, antibody isotype switching, and affinity maturation remain normal. This study demonstrates a new functional domain in CD40 that controls critical aspects of B-cell immunity in an in vivo setting.
Collapse
Affiliation(s)
- Li-Fan Lu
- Department of Microbiology and Immunology, Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, NH 03756, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Metastatic melanoma is an aggressive skin cancer that is notoriously resistant to current cancer therapies. In human melanoma, nuclear factor-kappa B (NF-kappaB) is upregulated, leading to the deregulation of gene transcription. In this review, we discuss (i) the relationship between gene alteration in melanoma and upregulation of NF-kappaB, (ii) mechanisms by which activated NF-kappaB switch from pro-apoptotic to anti-apoptotic functions in melanoma and (iii) autocrine mechanisms that promote constitutive activation of NF-kappaB in metastatic melanoma.
Collapse
Affiliation(s)
- Yukiko Ueda
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ann Richmond
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Veterans Affairs, Nashville, TN, USA
- Address correspondence to Ann Richmond, e-mail:
| |
Collapse
|
15
|
Hauer J, Püschner S, Ramakrishnan P, Simon U, Bongers M, Federle C, Engelmann H. TNF receptor (TNFR)-associated factor (TRAF) 3 serves as an inhibitor of TRAF2/5-mediated activation of the noncanonical NF-kappaB pathway by TRAF-binding TNFRs. Proc Natl Acad Sci U S A 2005; 102:2874-9. [PMID: 15708970 PMCID: PMC549490 DOI: 10.1073/pnas.0500187102] [Citation(s) in RCA: 193] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
TNF family members and their receptors contribute to increased gene expression for inflammatory processes and intracellular cascades leading to programmed cell death, both via activation of NF-kappaB. TNF receptor (TNFR)-associated factors (TRAFs) are cytoplasmic adaptor proteins binding to various receptors of the TNFR family. In an attempt to delineate the role of individual TRAFs, we compared NF-kappaB activation by CD40(wt) and CD40 mutants with different TRAF recruitment patterns. Recognized only recently, NF-kappaB signaling occurs at least via two different pathways. Each pathway results in nuclear translocation of two different Reldimers, the canonical p50/RelA and the noncanonical p52/RelB. Here, we show that via TRAF6, CD40 mediates only the activation of the canonical NF-kappaB pathway. Via TRAF2/5, CD40 activates both the canonical and the noncanonical NF-kappaB pathways. We observed that TRAF3 specifically blocked the NF-kappaB activation via TRAF2/5. This inhibitory effect of TRAF3 depends on the presence of an intact zinc finger domain. Paradoxically, suppression of TRAF2/5-mediated NF-kappaB activation by TRAF3 resulted in enhanced transcriptional activity of TRAF6-mediated canonical NF-kappaB emanating from CD40. We also observed that 12 TNFR family members (p75TNFR, LTbetaR, RANK, HVEM, CD40, CD30, CD27, 4-1BB, GITR, BCMA, OX40, and TACI) are each capable of activating the alternative NF-kappaB pathway and conclude that TRAF3 serves as a negative regulator of this pathway for all tested receptors.
Collapse
Affiliation(s)
- Julia Hauer
- Institut für Immunologie der Universität München, Goethestrasse 31, 80366 Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
16
|
He L, Grammer AC, Wu X, Lipsky PE. TRAF3 forms heterotrimers with TRAF2 and modulates its ability to mediate NF-{kappa}B activation. J Biol Chem 2004; 279:55855-65. [PMID: 15383523 DOI: 10.1074/jbc.m407284200] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
FRET experiments utilizing confocal microscopy or flow cytometry assessed homo- and heterotrimeric association of human tumor necrosis factor receptor-associated factors (TRAF) in living cells. Following transfection of HeLa cells with plasmids expressing CFP- or YFP-TRAF fusion proteins, constitutive homotypic association of TRAF2, -3, and -5 was observed, as well as heterotypic association of TRAF1-TRAF2 and TRAF3-TRAF5. A novel heterotypic association between TRAF2 and -3 was detected and confirmed by immunoprecipitation in Ramos B cells that constitutively express both TRAF2 and -3. Experiments employing deletion mutants of TRAF2 and TRAF3 revealed that this heterotypic interaction minimally involved the TRAF-C domain of TRAF3 as well as the TRAF-N domain and zinc fingers 4 and 5 of TRAF2. A novel flow cytometric FRET analysis utilizing a two-step approach to achieve linked FRET from CFP to YFP to HcRed established that TRAF2 and -3 constitutively form homo- and heterotrimers. The functional importance of TRAF2-TRAF3 heterotrimerization was demonstrated by the finding that TRAF3 inhibited spontaneous NF-kappaB, but not AP-1, activation induced by TRAF2. Ligation of CD40 on Ramos B cells by recombinant CD154 caused TRAF2 and TRAF3 to dissociate, whereas overexpression of TRAF3 in Ramos B cells inhibited CD154-induced TRAF2-mediated activation of NF-kappaB. Together, these results reveal a novel association between TRAF2 and TRAF3 that is mediated by unique portions of each protein and that specifically regulates activation of NF-kappaB, but not AP-1.
Collapse
Affiliation(s)
- Liusheng He
- Flow Cytometry Section in the Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases/NIH, 9000 Rockville Pike, Building 10, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
17
|
Rangaswami H, Bulbule A, Kundu GC. Nuclear factor-inducing kinase plays a crucial role in osteopontin-induced MAPK/IkappaBalpha kinase-dependent nuclear factor kappaB-mediated promatrix metalloproteinase-9 activation. J Biol Chem 2004; 279:38921-35. [PMID: 15247285 DOI: 10.1074/jbc.m404674200] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We have recently demonstrated that osteopontin (OPN) induces nuclear factor kappaB (NFkappaB)-mediated promatrix metalloproteinase-2 activation through IkappaBalpha/IkappaBalpha kinase (IKK) signaling pathways. However, the molecular mechanism(s) by which OPN regulates promatrix metalloproteinase-9 (pro-MMP-9) activation, MMP-9-dependent cell motility, and tumor growth and the involvement of upstream kinases in regulation of these processes in murine melanoma cells are not well defined. Here we report that OPN induced alpha(v)beta(3) integrin-mediated phosphorylation and activation of nuclear factor-inducing kinase (NIK) and enhanced the interaction between phosphorylated NIK and IKKalpha/beta in B16F10 cells. Moreover, NIK was involved in OPN-induced phosphorylations of MEK-1 and ERK1/2 in these cells. OPN induced NIK-dependent NFkappaB activation through ERK/IKKalpha/beta-mediated pathways. Furthermore OPN enhanced NIK-regulated urokinase-type plasminogen activator (uPA) secretion, uPA-dependent pro-MMP-9 activation, cell motility, and tumor growth. Wild type NIK, IKKalpha/beta, and ERK1/2 enhanced and kinase-negative NIK (mut NIK), dominant negative IKKalpha/beta (dn IKKalpha/beta), and dn ERK1/2 suppressed the OPN-induced NFkappaB activation, uPA secretion, pro-MMP-9 activation, cell motility, and chemoinvasion. Pretreatment of cells with anti-MMP-2 antibody along with anti-MMP-9 antibody drastically inhibited the OPN-induced cell migration and chemoinvasion, whereas cells pretreated with anti-MMP-2 antibody had no effect on OPN-induced pro-MMP-9 activation suggesting that OPN induces pro-MMP-2 and pro-MMP-9 activations through two distinct pathways. The level of active MMP-9 in the OPN-induced tumor was higher compared with control. To our knowledge, this is the first report that NIK plays a crucial role in OPN-induced NFkappaB activation, uPA secretion, and pro-MMP-9 activation through MAPK/IKKalpha/beta-mediated pathways, and all of these ultimately control the cell motility, invasiveness, and tumor growth.
Collapse
|
18
|
Kobayashi M, Takaori-Kondo A, Shindo K, Abudu A, Fukunaga K, Uchiyama T. APOBEC3G targets specific virus species. J Virol 2004; 78:8238-44. [PMID: 15254195 PMCID: PMC446120 DOI: 10.1128/jvi.78.15.8238-8244.2004] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human APOBEC3G (huAPOBEC3G), also known as CEM15, is a broad antiretroviral host factor that deaminates dC to dU in the minus strand DNA of human immunodeficiency virus type 1 (HIV-1), other lentiviruses, and murine leukemia virus (MLV), thereby creating G-to-A hypermutation in the plus strand DNA to inhibit the infectivity of these viruses. In this study, we examined the antiretroviral function of a murine homologue of APOBEC3G (muAPOBEC3G) on several retrovirus systems with different producer cells. MuAPOBEC3G did not suppress the infectivity of murine retroviral vectors produced from human or murine cells, whereas it showed antiviral activity on both wild-type and Deltavif virions of HIV-1 in human cells. In contrast, huAPOBEC3G showed broad antiviral activity on HIV-1 and murine retroviral vectors produced from human cells as well as murine cells. These data suggested that muAPOBEC3G does not possess antiretroviral activity on murine retroviruses and has a different target specificity from that of huAPOBEC3G and that huAPOBEC3G works as a broad antiviral factor not only in human cells but also in murine cells. A functional interaction study between human and murine APOBEC3G supported the former hypothesis. Furthermore, studies on the expression of APOBEC3G in producer cells and its incorporation into virions revealed that muAPOBEC3G is incorporated into HIV-1 virions but not into MLV virions. Thus, muAPOBEC3G cannot suppress the infectivity of murine retrovirus because it is not incorporated into virions. We suggest that murine retroviruses can replicate in murine target cells expressing muAPOBEC3G because they are not targets for this enzyme.
Collapse
Affiliation(s)
- Masayuki Kobayashi
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
19
|
Leyfer D, Bond A, Tworog E, Perron D, Maska S, Brito A, Kamens J, Weng Z, Voss J. cis-Element clustering correlates with dose-dependent pro- and antisignaling effects of IL18. Genes Immun 2004; 5:354-62. [PMID: 15190265 DOI: 10.1038/sj.gene.6364099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We examine the effects of IL18 on monocytes by performing microarray experiments using cell line KG1. Based on sensitivity to IL18, we identified three functionally distinct gene expression clusters (EC). We see little proinflammatory gene induction at low IL18 concentrations, but instead observe induction of diverse NF kappa B signaling inhibitors. Conversely, intermediate concentrations of IL18 induced proinflammatory genes including the activating subunits of NF kappa B. At the highest IL18 concentration, we observe a third gene cluster containing the proapoptotic Fas gene among others. Clustering of IL18-responsive genes based on cis-elements in their promoters agreed well with the ECs. We conclude that IL18 produces a dose-dependent transcriptional response that can in part be attributed to the composition of cis-elements in the promoters of IL18-responsive genes. These results also support a model for regulatory mechanisms that prevent spurious immune response due to weak cytokine fluctuations and a separate mechanism enabling induction of proinflammatory functions by higher levels of cytokine.
Collapse
Affiliation(s)
- D Leyfer
- Bioinformatics Program, Boston University, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Prell RA, Evans DE, Thalhofer C, Shi T, Funatake C, Weinberg AD. OX40-mediated memory T cell generation is TNF receptor-associated factor 2 dependent. THE JOURNAL OF IMMUNOLOGY 2004; 171:5997-6005. [PMID: 14634111 DOI: 10.4049/jimmunol.171.11.5997] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tumor necrosis factor receptor-associated factor 2 (TRAF2), an adapter protein that associates with the cytoplasmic tail of OX40, may play a critical role in OX40-mediated signal transduction. To investigate the in vivo role of TRAF2 in OX40-mediated generation of Ag-specific memory T cells, we bred OVA-specific TCR transgenic mice to TRAF2 dominant-negative (TRAF2 DN) mice. Following Ag stimulation and OX40 engagement of TRAF2 DN T cells in vivo, the number of long-lived OVA-specific T cells and effector T cell function was dramatically reduced when compared with wild-type T cells. We also demonstrate that CTLA-4 is down-regulated following OX40 engagement in vivo and the OX40-specific TRAF2 DN defect was partially overcome by CTLA-4 blockade in vivo. The data provide evidence that TRAF2 is linked to OX40-mediated memory T cell expansion and survival, and point to the down-regulation of CTLA-4 as a possible control element to enhance early T cell expansion through OX40 signaling.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, CD
- Antigens, Differentiation/biosynthesis
- Antigens, Differentiation/physiology
- CTLA-4 Antigen
- Cell Division/immunology
- Cells, Cultured
- Epitopes, T-Lymphocyte/immunology
- Growth Inhibitors/biosynthesis
- Growth Inhibitors/physiology
- Immune Sera/administration & dosage
- Immunologic Memory
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Lymphocyte Activation/immunology
- Lymphocyte Subsets/immunology
- Lymphocyte Subsets/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Oligonucleotide Array Sequence Analysis
- Ovalbumin/administration & dosage
- Ovalbumin/immunology
- Ovalbumin/pharmacology
- Protein Biosynthesis
- Proteins/genetics
- Proteins/physiology
- RNA/isolation & purification
- Receptors, OX40
- Receptors, Tumor Necrosis Factor/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor/physiology
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- TNF Receptor-Associated Factor 2
Collapse
Affiliation(s)
- Rodney A Prell
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Research Center, Providence Portland Medical Center, Portland, OR 97213, USA
| | | | | | | | | | | |
Collapse
|
21
|
Shindo K, Takaori-Kondo A, Kobayashi M, Abudu A, Fukunaga K, Uchiyama T. The enzymatic activity of CEM15/Apobec-3G is essential for the regulation of the infectivity of HIV-1 virion but not a sole determinant of its antiviral activity. J Biol Chem 2003; 278:44412-6. [PMID: 12970355 DOI: 10.1074/jbc.c300376200] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human immunodeficiency virus, type 1 (HIV-1) Vif protein plays an essential role in the regulation of the infectivity of HIV-1 virion. Vif functions to counteract an anti-HIV-1 cellular factor in non-permissive cells, CEM15/Apobec-3G, which shares a cytidine deaminase motif. CEM15/Apobec-3G deaminates dC to dU in the minus strand DNA of HIV-1, resulting in G to A hypermutation in the plus strand DNA. In this study, we have done the mutagenesis analysis on two cytidine deaminase motifs in CEM15/Apobec-3G and examined their antiviral functions as well as the DNA editing activity. Point mutations in the C-terminal active site such as E259Q and C291A almost completely abrogated the antiviral function, while those in the N-terminal active site such as E67Q and C100A retained this activity to a lesser extent as compared with that of the wild type. The DNA editing activities of E67Q and E259Q mutants were both retained but impaired to the same extent. This indicates that the enzymatic activity of this protein is essential but not a sole determinant of the antiviral activity. Furthermore, all the deletion mutants tested in this study lost the antiviral activity because of the loss of the activity for dimerization, suggesting that the entire protein structure is necessary for the antiviral function.
Collapse
Affiliation(s)
- Keisuke Shindo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaracho, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | |
Collapse
|
22
|
Abstract
TNF-receptor-associated factors (TRAFs) are the bottleneck of the TNF-receptor (TNF-R) family signal transduction. They integrate the signalling from many members of the TNF-R family and initiate intracellular signalling cascades aimed at the activation of NF-kappaB and c-jun, the reprogramming of gene expression and the control of cell death. Deregulation of these pathways is the cause of several autoimmune and inflammatory diseases. The specificity and interaction of the members of the TRAF family with the TNF-R entails the recognition of just a 4 - 6 amino acid motif in the cytosolic region of the receptor, suitable as an attractive target for drug discovery. This review summarises the current knowledge on TRAFs and discusses the pros and cons of their application as targets for drug discovery.
Collapse
Affiliation(s)
- Juan M Zapata
- The Burnham Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
23
|
Qian Y, Zhao Z, Jiang Z, Li X. Role of NF kappa B activator Act1 in CD40-mediated signaling in epithelial cells. Proc Natl Acad Sci U S A 2002; 99:9386-91. [PMID: 12089335 PMCID: PMC123150 DOI: 10.1073/pnas.142294499] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
CD40, a cell surface receptor in the tumor necrosis factor receptor family, first identified and functionally characterized on B lymphocytes, is also expressed on epithelial and other cells and is now thought to play a more general role in immune regulation. Overexpression of the NF kappa B activator 1 (Act1) leads to the activation of both NF kappa B and Jun kinase in epithelial cell lines. Endogenous Act1 is recruited to the CD40 receptor in human intestinal (HT29) and cervical (HeLa) epithelial cells upon stimulation with CD40 ligand, indicating that Act1 is involved in this signaling pathway. Act1 also interacts with tumor necrosis factor receptor-associated factor 3, a component involved in CD40-activated pathway. Furthermore, transfection of Act1 into C33A cervical epithelial cells, which do not express it, renders these cells sensitive to CD40 ligand-induced NF kappa B activation and protects them from CD40 ligand-induced apoptosis. We conclude that Act1 plays an important role in CD40-mediated signaling in epithelial cells.
Collapse
Affiliation(s)
- Youcun Qian
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
24
|
Affiliation(s)
- Punita Dhawan
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ann Richmond
- Department of Veterans Affairs, Nashville, Tennessee; and
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
25
|
Dhawan P, Richmond A. A novel NF-kappa B-inducing kinase-MAPK signaling pathway up-regulates NF-kappa B activity in melanoma cells. J Biol Chem 2002; 277:7920-8. [PMID: 11773061 PMCID: PMC2668260 DOI: 10.1074/jbc.m112210200] [Citation(s) in RCA: 228] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Constitutive activation of NF-kappa B is an emerging hallmark of various types of tumors including breast, colon, pancreatic, ovarian, and melanoma. In melanoma cells, the basal expression of the CXC chemokine, CXCL1, is constitutively up-regulated. This up-regulation can be attributed in part to constitutive activation of NF-kappa B. Previous studies have shown an elevated basal I kappa B kinase (IKK) activity in Hs294T melanoma cells, which leads to an increased rate of I kappa B phosphorylation and degradation. This increase in I kappa B-alpha phosphorylation and degradation leads to an approximately 19-fold higher nuclear localization of NF-kappa B. However, the upstream IKK kinase activity is up-regulated by only about 2-fold and cannot account for the observed increase in NF-kappa B activity. We now demonstrate that NF-kappa B-inducing kinase (NIK) is highly expressed in melanoma cells, and IKK-associated NIK activity is enhanced in these cells compared with the normal cells. Kinase-dead NIK blocked constitutive NF-kappa B or CXCL1 promoter activity in Hs294T melanoma cells, but not in control normal human epidermal melanocytes. Transient overexpression of wild type NIK results in increased phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), which is inhibited in a concentration-dependent manner by PD98059, an inhibitor of p42/44 MAPK. Moreover, the NF-kappa B promoter activity decreased with overexpression of dominant negative ERK expression constructs, and EMSA analyses further support the hypothesis that ERK acts upstream of NF-kappa B and regulates the NF-kappa B DNA binding activity. Taken together, our data implicate involvement of I kappa B kinase and MAPK signaling cascades in NIK-induced constitutive activation of NF-kappa B.
Collapse
Affiliation(s)
- Punita Dhawan
- Department of Veterans Affairs, Nashville, Tennessee 37212
| | - Ann Richmond
- Department of Veterans Affairs, Nashville, Tennessee 37212
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
- To whom correspondence should be addressed: Dept. of Cancer Biology, Vanderbilt University School of Medicine, MCN T-2212, Nashville, TN 37232. Tel.: 615-343-7777; Fax: 615-343-4539; E-mail:
| |
Collapse
|
26
|
Chung JY, Park YC, Ye H, Wu H. All TRAFs are not created equal: common and distinct molecular mechanisms of TRAF-mediated signal transduction. J Cell Sci 2002; 115:679-88. [PMID: 11865024 DOI: 10.1242/jcs.115.4.679] [Citation(s) in RCA: 423] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The tumor necrosis factor (TNF) receptor associated factors (TRAFs) have emerged as the major signal transducers for the TNF receptor superfamily and the interleukin-1 receptor/Toll-like receptor (IL-1R/TLR) superfamily. TRAFs collectively play important functions in both adaptive and innate immunity. Recent functional and structural studies have revealed the individuality of each of the mammalian TRAFs and advanced our understanding of the underlying molecular mechanisms. Here, we examine this functional divergence among TRAFs from a perspective of both upstream and downstream TRAF signal transduction pathways and of signaling-dependent regulation of TRAF trafficking. We raise additional questions and propose hypotheses regarding the molecular basis of TRAF signaling specificity.
Collapse
Affiliation(s)
- Jee Y Chung
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | |
Collapse
|