Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 16(12): e1008504, 2020 12.
Article in English | MEDLINE | ID: mdl-33362245

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia/lymphoma (ATLL), and the neurological disease HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). The HTLV-1 Tax protein persistently activates the NF-κB pathway to enhance the proliferation and survival of HTLV-1 infected T cells. Lysine 63 (K63)-linked polyubiquitination of Tax provides an important regulatory mechanism that promotes Tax-mediated interaction with the IKK complex and activation of NF-κB; however, the host proteins regulating Tax ubiquitination are largely unknown. To identify new Tax interacting proteins that may regulate its ubiquitination we conducted a yeast two-hybrid screen using Tax as bait. This screen yielded the E3/E4 ubiquitin conjugation factor UBE4B as a novel binding partner for Tax. Here, we confirmed the interaction between Tax and UBE4B in mammalian cells by co-immunoprecipitation assays and demonstrated colocalization by proximity ligation assay and confocal microscopy. Overexpression of UBE4B specifically enhanced Tax-induced NF-κB activation, whereas knockdown of UBE4B impaired Tax-induced NF-κB activation and the induction of NF-κB target genes in T cells and ATLL cell lines. Furthermore, depletion of UBE4B with shRNA resulted in apoptotic cell death and diminished the proliferation of ATLL cell lines. Finally, overexpression of UBE4B enhanced Tax polyubiquitination, and knockdown or CRISPR/Cas9-mediated knockout of UBE4B attenuated both K48- and K63-linked polyubiquitination of Tax. Collectively, these results implicate UBE4B in HTLV-1 Tax polyubiquitination and downstream NF-κB activation.


Subject(s)
Gene Products, tax/metabolism , NF-kappa B/metabolism , Ubiquitin-Protein Ligases/metabolism , Gene Expression Regulation/genetics , Genes, pX/physiology , HEK293 Cells , Human T-lymphotropic virus 1/metabolism , Human T-lymphotropic virus 1/pathogenicity , Humans , NF-kappa B/physiology , Signal Transduction/physiology , T-Lymphocytes/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitination , Ubiquitins/metabolism
2.
J Biol Chem ; 290(23): 14729-39, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25911105

ABSTRACT

The transcription factor IRF7 (interferon regulatory factor 7) is a key regulator of type I interferon and plays essential roles in restricting virus infection and spread. IRF7 activation is tightly regulated to prevent excessive inflammation and autoimmunity; however, how IRF7 is suppressed by negative regulators remains poorly understood. Here, we have identified AIP (aryl hydrocarbon receptor interacting protein) as a new binding partner of IRF7. The interaction between AIP and IRF7 is enhanced upon virus infection, and AIP potently inhibits IRF7-induced type I IFN (IFNα/ß) production. Overexpression of AIP blocks virus-induced activation of IFN, whereas knockdown of AIP by siRNA potentiates virally activated IFN production. Consistently, AIP-deficient murine embryonic fibroblasts are highly resistant to virus infection because of increased production of IFNα/ß. AIP inhibits IRF7 function by antagonizing the nuclear localization of IRF7. Together, our study identifies AIP as a novel inhibitor of IRF7 and a negative regulator of innate antiviral signaling.


Subject(s)
Interferon Regulatory Factor-7/immunology , Interferon-alpha/immunology , Interferon-beta/immunology , Intracellular Signaling Peptides and Proteins/immunology , Virus Diseases/immunology , Animals , Cell Line , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , RNA Interference , RNA, Small Interfering/genetics , Up-Regulation , Virus Diseases/genetics
3.
Retrovirology ; 12: 23, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25809782

ABSTRACT

BACKGROUND: The exact molecular mechanisms regarding HTLV-1 Tax-mediated viral gene expression and CD4 T-cell transformation have yet to be fully delineated. Herein, utilizing virus-infected primary CD4+ T cells and the virus-producing cell line, MT-2, we describe the involvement and regulation of Myocyte enhancer factor-2 (specifically MEF-2A) during the course of HTLV-1 infection and associated disease syndrome. RESULTS: Inhibition of MEF-2 expression by shRNA and its activity by HDAC9 led to reduced viral replication and T-cell transformation in correlation with a heightened expression of MEF-2 in ATL patients. Mechanistically, MEF-2 was recruited to the viral promoter (LTR, long terminal repeat) in the context of chromatin, and constituted Tax/CREB transcriptional complex via direct binding to the HTLV-1 LTR. Furthermore, an increase in MEF-2 expression was observed upon infection in an extent similar to CREB (known Tax-interacting transcription factor), and HATs (p300, CBP, and p/CAF). Confocal imaging confirmed MEF-2 co-localization with Tax and these proteins were also shown to interact by co-immunoprecipitation. MEF-2 stabilization of Tax/CREB complex was confirmed by a novel promoter-binding assay that highlighted the involvement of NFAT (nuclear factor of activated T cells) in this process via Tax-mediated activation of calcineurin (a calcium-dependent serine-threonine phosphatase). MEF-2-integrated signaling pathways (PI3K/Akt, NF-κB, MAPK, JAK/STAT, and TGF-ß) were also activated during HTLV-1 infection of primary CD4+ T cells, possibly regulating MEF-2 activity. CONCLUSIONS: We demonstrate the involvement of MEF-2 in Tax-mediated LTR activation, viral replication, and T-cell transformation in correlation with its heightened expression in ATL patients through direct binding to DNA within the HTLV-1 LTR.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Transformation, Viral , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Products, tax/metabolism , Host-Pathogen Interactions , Human T-lymphotropic virus 1/physiology , CD4-Positive T-Lymphocytes/physiology , Cells, Cultured , Humans , MEF2 Transcription Factors/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Multimerization , Transcription, Genetic , Virus Replication
4.
J Virol ; 89(9): 4880-93, 2015 May.
Article in English | MEDLINE | ID: mdl-25694597

ABSTRACT

UNLABELLED: Human T-cell lymphotropic virus type I (HTLV-1) is an oncogenic retrovirus considered to be the etiological agent of adult T-cell leukemia (ATL). The viral transactivator Tax is regarded as the oncoprotein responsible for contributing toward the transformation process. Here, we demonstrate that Tax potently inhibits the activity of DEx(D/H) box helicases RIG-I and MDA5 as well as Toll-dependent TIR-domain-containing adapter-inducing interferon-ß (TRIF), which function as cellular sensors or mediators of viral RNA and facilitate innate immune responses, including the production of type I IFN. Tax manifested this function by binding to the RIP homotypic interaction motif (RHIM) domains of TRIF and RIP1 to disrupt interferon regulatory factor 7 (IRF7) activity, a critical type I IFN transcription factor. These data provide further mechanistic insight into HTLV-1-mediated subversion of cellular host defense responses, which may help explain HTLV-1-related pathogenesis and oncogenesis. IMPORTANCE: It is predicted that up to 15% of all human cancers may involve virus infection. For example, human T-cell lymphotropic virus type 1 (HTLV-1) has been reported to infect up to 25 million people worldwide and is the causative agent of adult T-cell leukemia (ATL). We show here that HTLV-1 may be able to successfully infect the T cells and remain latent due to the virally encoded product Tax inhibiting a key host defense pathway. Understanding the mechanisms by which Tax subverts the immune system may lead to the development of a therapeutic treatment for HTLV-1-mediated disease.


Subject(s)
Gene Products, tax/metabolism , Human T-lymphotropic virus 1/physiology , Immune Evasion , Immune Tolerance , Immunity, Innate , Signal Transduction , Animals , Cell Line , Host-Pathogen Interactions , Humans , Mice, Inbred BALB C
5.
J Immunol ; 194(7): 3286-94, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25710910

ABSTRACT

IL-17 is a proinflammatory cytokine that promotes the expression of different cytokines and chemokines via the induction of gene transcription and the posttranscriptional stabilization of mRNAs. In this study, we show that IL-17 increases the half-life of the Zc3h12a mRNA via interaction of the adaptor protein CIKS with the DEAD box protein DDX3X. IL-17 stimulation promotes the formation of a complex between CIKS and DDX3X, and this interaction requires the helicase domain of DDX3X but not its ATPase activity. DDX3X knockdown decreases the IL-17-induced stability of Zc3h12a without affecting the stability of other mRNAs. IKKε, TNFR-associated factor 2, and TNFR-associated factor 5 were also required to mediate the IL-17-induced Zc3h12a stabilization. DDX3X directly binds the Zc3h12a mRNA after IL-17 stimulation. Collectively, our findings define a novel, IL-17-dependent mechanism regulating the stabilization of a selected mRNA.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gene Expression Regulation , Interleukin-17/metabolism , RNA Stability , Ribonucleases/genetics , Transcription Factors/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Adaptor Proteins, Signal Transducing , Gene Expression Regulation/drug effects , Humans , I-kappa B Kinase/metabolism , Interleukin-17/pharmacology , Multiprotein Complexes/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 5/metabolism
6.
Viruses ; 6(10): 3925-43, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25341660

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is a complex retrovirus that infects CD4+ T cells and causes adult T-cell leukemia/lymphoma (ATLL) in 3%-5% of infected individuals after a long latent period. HTLV-1 Tax is a trans-activating protein that regulates viral gene expression and also modulates cellular signaling pathways to enhance T-cell proliferation and cell survival. The Tax oncoprotein promotes T-cell transformation, in part via constitutive activation of the NF-κB transcription factor; however, the underlying mechanisms remain unknown. Ubiquitination is a type of post-translational modification that occurs in a three-step enzymatic cascade mediated by E1, E2 and E3 enzymes and regulates protein stability as well as signal transduction, protein trafficking and the DNA damage response. Emerging studies indicate that Tax hijacks the ubiquitin machinery to activate ubiquitin-dependent kinases and downstream NF-κB signaling. Tax interacts with the E2 conjugating enzyme Ubc13 and is conjugated on C-terminal lysine residues with lysine 63-linked polyubiquitin chains. Tax K63-linked polyubiquitination may serve as a platform for signaling complexes since this modification is critical for interactions with NEMO and IKK. In addition to NF-κB signaling, mono- and polyubiquitination of Tax also regulate its subcellular trafficking and stability. Here, we review recent advances in the diverse roles of ubiquitin in Tax function and how Tax usurps the ubiquitin-proteasome pathway to promote oncogenesis.


Subject(s)
Gene Products, tax/metabolism , Human T-lymphotropic virus 1/metabolism , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Ubiquitin/metabolism , Carcinogenesis , Cell Line , Gene Expression Regulation , Gene Products, tax/genetics , Human T-lymphotropic virus 1/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/virology , Protein Transport , Signal Transduction , Ubiquitination
7.
PLoS Pathog ; 10(10): e1004418, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25340344

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) infection is linked to the development of adult T-cell leukemia (ATL) and the neuroinflammatory disease HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP). The HTLV-1 Tax protein functions as a potent viral oncogene that constitutively activates the NF-κB transcription factor to transform T cells; however, the underlying mechanisms remain obscure. Here, using next-generation RNA sequencing we identified the IL-25 receptor subunit IL-17RB as an aberrantly overexpressed gene in HTLV-1 immortalized T cells. Tax induced the expression of IL-17RB in an IκB kinase (IKK) and NF-κB-dependent manner. Remarkably, Tax activation of the canonical NF-κB pathway in T cells was critically dependent on IL-17RB expression. IL-17RB and IL-25 were required for HTLV-1-induced immortalization of primary T cells, and the constitutive NF-κB activation and survival of HTLV-1 transformed T cells. IL-9 was identified as an important downstream target gene of the IL-17RB pathway that drives the proliferation of HTLV-1 transformed cells. Furthermore, IL-17RB was overexpressed in leukemic cells from a subset of ATL patients and also regulated NF-κB activation in some, but not all, Tax-negative ATL cell lines. Together, our results support a model whereby Tax instigates an IL-17RB-NF-κB feed-forward autocrine loop that is obligatory for HTLV-1 leukemogenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Products, tax/metabolism , Human T-lymphotropic virus 1/metabolism , Interleukin-17/metabolism , NF-kappa B/metabolism , Signal Transduction , T-Lymphocytes/cytology , Gene Expression Regulation/physiology , Humans , Receptors, Interleukin-17/metabolism , Signal Transduction/physiology
8.
Future Virol ; 8(3): 223-227, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23730325

ABSTRACT

It is well established that the human T-cell leukemia virus type 1-encoded oncoprotein Tax (Tax1) undergoes polyubiquitination as part of its mechanism to persistently activate NF-κB. However, it remains unclear whether Tax2 encoded by the closely related human T-cell leukemia virus type 2 utilizes any post-translational mechanisms to activate NF-κB. This study examines the role of ubiquitination and SUMOylation in Tax2 activation of NF-κB. The authors have demonstrated that, in contrast to Tax1, Tax2 is not conjugated by ubiquitin or SUMO proteins. Overexpression of the E2 ubiquitin-conjugating enzyme Ubc13 specifically enhances Tax1, but not Tax2, ubiquitination and NF-κB activation. Furthermore, a Tax2 lysineless mutant that is unable to be ubiquitinated, SUMOylated or acetylated retains NEMO/IKKγ interactions and activation of the NF-κB pathway. Together, these results provide evidence that Tax1 and Tax2 utilize distinct mechanisms to activate NF-κB.

10.
J Virol ; 86(6): 3357-69, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22258247

ABSTRACT

The human T-cell leukemia virus type 1 (HTLV-1) Tax oncoprotein actively shuttles between the nucleus, where it interacts with transcriptional and splicing regulatory proteins, and the cytoplasm, where it activates NF-κB. Posttranslational modifications of Tax such as ubiquitination regulate its subcellular localization and hence its function; however, the regulation of Tax trafficking and NF-κB activation by host factors is poorly understood. By screening a deubiquitinating (DUB) enzyme small interfering RNA (siRNA) library, we identified the metalloprotease STAM-binding protein-like 1 (STAMBPL1) as a positive regulator of Tax-mediated NF-κB activation. Overexpression of wild-type STAMBPL1, but not a catalytically inactive mutant, enhanced Tax-mediated NF-κB activation, whereas silencing of STAMBPL1 with siRNA impaired Tax activation of both the canonical and noncanonical NF-κB signaling pathways. STAMBPL1 regulated Tax-induced NF-κB signaling indirectly by controlling Tax nuclear/cytoplasmic transport and was required for DNA damage-induced Tax nuclear export. Together, these results reveal that the deubiquitinase STAMBPL1 is a key regulator of Tax trafficking and function.


Subject(s)
Cell Nucleus/metabolism , Gene Products, tax/metabolism , HTLV-I Infections/enzymology , HTLV-I Infections/genetics , Human T-lymphotropic virus 1/metabolism , NF-kappa B/metabolism , RNA Interference , Ubiquitin Thiolesterase/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/genetics , Gene Expression Regulation , Gene Products, tax/genetics , HTLV-I Infections/virology , Human T-lymphotropic virus 1/genetics , Humans , NF-kappa B/genetics , Peptide Hydrolases , Ubiquitin Thiolesterase/genetics
11.
BMC Microbiol ; 10: 9, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20070893

ABSTRACT

BACKGROUND: The Pseudomonas aeruginosa major constitutive outer membrane porin protein F (OprF) has been shown to be a protective antigen and was previously used to activate an immunological response in a mouse model of lung pneumonia. The purpose of our study was to demonstrate the ability of mouse dendritic cells pulsed with purified or recombinant OprF to protect mice against P. aeruginosa infection and inflammation.Both native (n-OprF), isolated and purified from PAO1 bacterial strain, and recombinant (histidin-conjugated) OprF (His-OprF), obtained by cloning of the oprF gene into the pET28a expression vector, were used to stimulate dendritic cells in vitro before adoptive transfer into prospective recipient mice with P. aeruginosa pulmonary infection. RESULTS: Similar to n-OprF, His-OprF activated dendritic cells in vitro, inducing the costimulatory molecule expression as well as cytokine production. Upon adoptive transfer in vivo, porin-pulsed dendritic cells (DCs) induced Th1-mediated resistance to infection and associated inflammatory pathology caused by either the PAO1 strain or a clinically-isolated mucoid strain. CONCLUSIONS: This study highlights the pivotal contribution of DCs to vaccine-induced protection against P. aeruginosa infection and associated inflammation.


Subject(s)
Adoptive Transfer , Dendritic Cells/immunology , Porins/immunology , Pseudomonas Infections/prevention & control , Pseudomonas Vaccines/immunology , Pseudomonas aeruginosa/immunology , Animals , Female , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pseudomonas Infections/immunology , Th1 Cells/immunology
12.
Mol Immunol ; 46(16): 3278-82, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19698991

ABSTRACT

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


Subject(s)
B-Cell Activation Factor Receptor/biosynthesis , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 1/biosynthesis , TNF Receptor-Associated Factor 3/metabolism , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/immunology , Cell Line, Tumor , Gene Expression , Hodgkin Disease/genetics , Hodgkin Disease/immunology , Hodgkin Disease/metabolism , Humans , NF-kappa B/genetics , NF-kappa B/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Stability , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/immunology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , Zinc Fingers/genetics , Zinc Fingers/immunology , NF-kappaB-Inducing Kinase
13.
J Cell Physiol ; 220(2): 410-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19365808

ABSTRACT

NEMO/IKKgamma is the essential regulatory subunit of the IkB Kinase (IKK) complex, required for the activation of Nuclear Factor kB (NF-kB) in many physiological processes such as inflammation, immunity, apoptosis, or development. NEMO works at a converging point of the NF-kB pathway as it interacts with upstream signaling molecules to orchestrate its activation. Here we report on the identification of a novel NEMO-interacting protein, NESCA, an adapter molecule previously shown to be involved in the NGF-pathway via the TrkA receptor. We demonstrated that NESCA and NEMO interact by their N-terminal region. Beside to NEMO, we revealed that NESCA directly associates to the E3 ubiquitin ligase TRAF6, which in turn catalyzes NESCA polyubiquitination. Finally, we demonstrated that NESCA overexpression strongly inhibits TRAF6-mediated polyubiquitination of NEMO. In summary, our results highlight that NESCA represents a novel missing link in the NEMO-mediated NF-kB activation pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , I-kappa B Kinase/metabolism , TNF Receptor-Associated Factor 6/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Humans , I-kappa B Kinase/genetics , Polyubiquitin/metabolism , Protein Binding , TNF Receptor-Associated Factor 6/genetics , Two-Hybrid System Techniques , Ubiquitination
14.
Proc Natl Acad Sci U S A ; 105(37): 14058-63, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18768801

ABSTRACT

NF-kappaB is constitutively activated in primary human thyroid tumors, particularly in those of anaplastic type. The inhibition of NF-kappaB activity in the human anaplastic thyroid carcinoma cell line, FRO, leads to an increased susceptibility to chemotherapeutic drug-induced apoptosis and to the blockage of their ability to form tumors in nude mice. To identify NF-kappaB target genes involved in thyroid cancer, we analyzed the secretome of conditioned media from parental and NF-kappaB-null FRO cells. Proteomic analysis revealed that the neutrophil gelatinase-associated lipocalin (NGAL), a protein involved in inflammatory and immune responses, is secreted by FRO cells whereas its expression is strongly reduced in the NF-kappaB-null FRO cells. NGAL is highly expressed in human thyroid carcinomas, and knocking down its expression blocks the ability of FRO cells to grow in soft agar and form tumors in nude mice. These effects are reverted by the addition of either recombinant NGAL or FRO conditioned medium. In addition, we show that the prosurvival activity of NGAL is mediated by its ability to bind and transport iron inside the cells. Our data suggest that NF-kappaB contributes to thyroid tumor cell survival by controlling iron uptake via NGAL.


Subject(s)
Acute-Phase Proteins/metabolism , Gene Expression Regulation, Neoplastic , Lipocalins/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology , Acute-Phase Proteins/genetics , Cell Line, Tumor , Cell Survival , Health , Humans , I-kappa B Kinase/metabolism , Immunohistochemistry , Lipocalin-2 , Lipocalins/genetics , Proteomics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Thyroid Neoplasms/genetics
15.
J Biol Chem ; 281(27): 18482-8, 2006 Jul 07.
Article in English | MEDLINE | ID: mdl-16684768

ABSTRACT

Nuclear factor kappaB (NF-kappaB) plays a pivotal role in inflammation, immunity, stress responses, and protection from apoptosis. Canonical activation of NF-kappaB is dependent on the phosphorylation of the inhibitory subunit IkappaBalpha that is mediated by a multimeric, high molecular weight complex, called IkappaB kinase (IKK) complex. This is composed of two catalytic subunits, IKKalpha and IKKbeta, and a regulatory subunit, NEMO/IKKgamma. The latter protein is essential for the activation of IKKs and NF-kappaB, but its mechanism of action is not well understood. Here we identified ABIN-1 (A20 binding inhibitor of NF-kappaB) as a NEMO/IKKgamma-interacting protein. ABIN-1 has been previously identified as an A20-binding protein and it has been proposed to mediate the NF-kappaB inhibiting effects of A20. We find that both ABIN-1 and A20 inhibit NF-kappaB at the level of the IKK complex and that A20 inhibits activation of NF-kappaB by de-ubiquitination of NEMO/IKKgamma. Importantly, small interfering RNA targeting ABIN-1 abrogates A20-dependent de-ubiquitination of NEMO/IKKgamma and RNA interference of A20 impairs the ability of ABIN-1 to inhibit NF-kappaB activation. Altogether our data indicate that ABIN-1 physically links A20 to NEMO/IKKgamma and facilitates A20-mediated de-ubiquitination of NEMO/IKKgamma, thus resulting in inhibition of NF-kappaB.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Proteins/metabolism , Animals , Binding Sites , Catalytic Domain/genetics , Cell Line , Cysteine Endopeptidases , DNA-Binding Proteins/genetics , Humans , I-kappa B Kinase/genetics , Intracellular Signaling Peptides and Proteins , Mice , Mutation , NF-kappa B/antagonists & inhibitors , Nuclear Proteins , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary/genetics , Proteins/genetics , Rabbits , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin
SELECTION OF CITATIONS
SEARCH DETAIL
...