52 results on '"Gohda J"'
Search Results
2. PP 1.6 - Genome-wide RNAi screen identifies MAPK-RPK required for HIV-1 proviral silencing in non-T cell reservoir cell-line model
- Author
-
Takeuchi, H., Ishida, T., Satou, Y., Gohda, J., Kitamura, H., Gan, S., Takahashi, K., and Yamaoka, S.
- Published
- 2019
- Full Text
- View/download PDF
3. Activation of the I B kinase complex by HTLV-1 Tax requires cytosolic factors involved in Tax-induced polyubiquitination
- Author
-
Shibata, Y., primary, Tanaka, Y., additional, Gohda, J., additional, and Inoue, J.-i., additional
- Published
- 2011
- Full Text
- View/download PDF
4. In Silico and In Vitro Evaluation of Some Amidine Derivatives as Hit Compounds towards Development of Inhibitors against Coronavirus Diseases.
- Author
-
Hassan AHE, El-Sayed SM, Yamamoto M, Gohda J, Matsumoto T, Shirouzu M, Inoue JI, Kawaguchi Y, Mansour RMA, Anvari A, and Farahat AA
- Subjects
- Humans, SARS-CoV-2, Benzamidines pharmacology, Virus Internalization, Antiviral Agents pharmacology, Antiviral Agents chemistry, COVID-19, Middle East Respiratory Syndrome Coronavirus
- Abstract
Coronaviruses, including SARS-CoV-2, SARS-CoV, MERS-CoV and influenza A virus, require the host proteases to mediate viral entry into cells. Rather than targeting the continuously mutating viral proteins, targeting the conserved host-based entry mechanism could offer advantages. Nafamostat and camostat were discovered as covalent inhibitors of TMPRSS2 protease involved in viral entry. To circumvent their limitations, a reversible inhibitor might be required. Considering nafamostat structure and using pentamidine as a starting point, a small set of structurally diverse rigid analogues were designed and evaluated in silico to guide selection of compounds to be prepared for biological evaluation. Based on the results of in silico study, six compounds were prepared and evaluated in vitro. At the enzyme level, compounds 10 - 12 triggered potential TMPRSS2 inhibition with low micromolar IC
50 concentrations, but they were less effective in cellular assays. Meanwhile, compound 14 did not trigger potential TMPRSS2 inhibition at the enzyme level, but it showed potential cellular activity regarding inhibition of membrane fusion with a low micromolar IC50 value of 10.87 µM, suggesting its action could be mediated by another molecular target. Furthermore, in vitro evaluation showed that compound 14 inhibited pseudovirus entry as well as thrombin and factor Xa. Together, this study presents compound 14 as a hit compound that might serve as a starting point for developing potential viral entry inhibitors with possible application against coronaviruses.- Published
- 2023
- Full Text
- View/download PDF
5. Interferon regulatory factor 3 mediates effective antiviral responses to human coronavirus 229E and OC43 infection.
- Author
-
Duncan JKS, Xu D, Licursi M, Joyce MA, Saffran HA, Liu K, Gohda J, Tyrrell DL, Kawaguchi Y, and Hirasawa K
- Subjects
- Humans, Interferon Regulatory Factor-3, SARS-CoV-2 metabolism, Interferons metabolism, Antiviral Agents pharmacology, Interferon Regulatory Factors, Coronavirus 229E, Human, COVID-19
- Abstract
Interferon regulatory factors (IRFs) are key elements of antiviral innate responses that regulate the transcription of interferons (IFNs) and IFN-stimulated genes (ISGs). While the sensitivity of human coronaviruses to IFNs has been characterized, antiviral roles of IRFs during human coronavirus infection are not fully understood. Type I or II IFN treatment protected MRC5 cells from human coronavirus 229E infection, but not OC43. Cells infected with 229E or OC43 upregulated ISGs, indicating that antiviral transcription is not suppressed. Antiviral IRFs, IRF1, IRF3 and IRF7, were activated in cells infected with 229E, OC43 or severe acute respiratory syndrome-associated coronavirus 2 (SARS-CoV-2). RNAi knockdown and overexpression of IRFs demonstrated that IRF1 and IRF3 have antiviral properties against OC43, while IRF3 and IRF7 are effective in restricting 229E infection. IRF3 activation effectively promotes transcription of antiviral genes during OC43 or 229E infection. Our study suggests that IRFs may be effective antiviral regulators against human coronavirus infection., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Duncan, Xu, Licursi, Joyce, Saffran, Liu, Gohda, Tyrrell, Kawaguchi and Hirasawa.)
- Published
- 2023
- Full Text
- View/download PDF
6. Reactive oxygen species are associated with the inhibitory effect of N-(4-hydroxyphenyl)-retinamide on the entry of the severe acute respiratory syndrome-coronavirus 2.
- Author
-
Hayashi Y, Huang X, Tanikawa T, Tanigawa K, Yamamoto M, Gohda J, Inoue JI, Fukase K, and Kabayama K
- Subjects
- Humans, Reactive Oxygen Species metabolism, SARS-CoV-2 metabolism, Apoptosis, Oxidoreductases, Fenretinide pharmacology, Antineoplastic Agents pharmacology, COVID-19
- Abstract
N-(4-hydroxyphenyl)-retinamide (4-HPR) inhibits the dihydroceramide Δ4-desaturase 1 (DEGS1) enzymatic activity. We previously reported that 4-HPR suppresses the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) spike protein-mediated membrane fusion through a decrease in membrane fluidity in a DEGS1-independent manner. However, the precise mechanism underlying the inhibition of viral entry by 4-HPR remains unclear. In this study, we examined the role of reactive oxygen species (ROS) in the inhibition of membrane fusion by 4-HPR because 4-HPR is a well-known ROS-inducing agent. Intracellular ROS generation was found to be increased in the target cells in a cell-cell fusion assay after 4-HPR treatment, which was attenuated by the addition of the antioxidant, α-tocopherol (TCP). The reduction in membrane fusion susceptibility by 4-HPR treatment in the cell-cell fusion assay was alleviated by TCP addition. Furthermore, fluorescence recovery after photobleaching analysis showed that the lateral diffusion of glycosylphosphatidylinositol-anchored protein and SARS CoV-2 receptor was reduced by 4-HPR treatment and restored by TCP addition. These results indicate that the decrease in SARS-CoV-2 spike protein-mediated membrane fusion and membrane fluidity by 4-HPR was due to ROS generation. Taken together, these results demonstrate that ROS production is associated with the 4-HPR inhibitory effect on SARS-CoV-2 entry., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Japanese Biochemical Society. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
7. Dysfunctional Sars-CoV-2-M protein-specific cytotoxic T lymphocytes in patients recovering from severe COVID-19.
- Author
-
Ogura H, Gohda J, Lu X, Yamamoto M, Takesue Y, Son A, Doi S, Matsushita K, Isobe F, Fukuda Y, Huang TP, Ueno T, Mambo N, Murakami H, Kawaguchi Y, Inoue JI, Shirai K, Yamasaki S, Hirata JI, and Ishido S
- Subjects
- Humans, SARS-CoV-2, T-Lymphocytes, Cytotoxic, Epitopes, T-Lymphocyte, HLA-A Antigens, CD8-Positive T-Lymphocytes, COVID-19
- Abstract
Although the importance of virus-specific cytotoxic T lymphocytes (CTL) in virus clearance is evident in COVID-19, the characteristics of virus-specific CTLs related to disease severity have not been fully explored. Here we show that the phenotype of virus-specific CTLs against immunoprevalent epitopes in COVID-19 convalescents might differ according to the course of the disease. We establish a cellular screening method that uses artificial antigen presenting cells, expressing HLA-A
* 24:02, the costimulatory molecule 4-1BBL, SARS-CoV-2 structural proteins S, M, and N and non-structural proteins ORF3a and nsp6/ORF1a. The screen implicates SARS-CoV-2 M protein as a frequent target of IFNγ secreting CD8+ T cells, and identifies M198-206 as an immunoprevalent epitope in our cohort of HLA-A* 24:02 positive convalescent COVID-19 patients recovering from mild, moderate and severe disease. Further exploration of M198-206 -specific CD8+ T cells with single cell RNA sequencing reveals public TCRs in virus-specific CD8+ T cells, and shows an exhausted phenotype with less differentiated status in cells from the severe group compared to cells from the moderate group. In summary, this study describes a method to identify T cell epitopes, indicate that dysfunction of virus-specific CTLs might be an important determinant of clinical outcomes., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
8. Metalloproteinase-Dependent and TMPRSS2-Independent Cell Surface Entry Pathway of SARS-CoV-2 Requires the Furin Cleavage Site and the S2 Domain of Spike Protein.
- Author
-
Yamamoto M, Gohda J, Kobayashi A, Tomita K, Hirayama Y, Koshikawa N, Seiki M, Semba K, Akiyama T, Kawaguchi Y, and Inoue JI
- Subjects
- Humans, Metalloproteases genetics, Serine Endopeptidases genetics, COVID-19, SARS-CoV-2 metabolism, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Virus Internalization
- Abstract
The ongoing global vaccination program to prevent SARS-CoV-2 infection, the causative agent of COVID-19, has had significant success. However, recently, virus variants that can evade the immunity in a host achieved through vaccination have emerged. Consequently, new therapeutic agents that can efficiently prevent infection from these new variants, and hence COVID-19 spread, are urgently required. To achieve this, extensive characterization of virus-host cell interactions to identify effective therapeutic targets is warranted. Here, we report a cell surface entry pathway of SARS-CoV-2 that exists in a cell type-dependent manner and is TMPRSS2 independent but sensitive to various broad-spectrum metalloproteinase inhibitors such as marimastat and prinomastat. Experiments with selective metalloproteinase inhibitors and gene-specific small interfering RNAS (siRNAs) revealed that a disintegrin and metalloproteinase 10 (ADAM10) is partially involved in the metalloproteinase pathway. Consistent with our finding that the pathway is unique to SARS-CoV-2 among highly pathogenic human coronaviruses, both the furin cleavage motif in the S1/S2 boundary and the S2 domain of SARS-CoV-2 spike protein are essential for metalloproteinase-dependent entry. In contrast, the two elements of SARS-CoV-2 independently contributed to TMPRSS2-dependent S2 priming. The metalloproteinase pathway is involved in SARS-CoV-2-induced syncytium formation and cytopathicity, leading us to theorize that it is also involved in the rapid spread of SARS-CoV-2 and the pathogenesis of COVID-19. Thus, targeting the metalloproteinase pathway in addition to the TMPRSS2 and endosomal pathways could be an effective strategy by which to cure COVID-19 in the future. IMPORTANCE To develop effective therapeutics against COVID-19, it is necessary to elucidate in detail the infection mechanism of the causative agent, SARS-CoV-2. SARS-CoV-2 binds to the cell surface receptor ACE2 via the spike protein, and then the spike protein is cleaved by host proteases to enable entry. Here, we found that the metalloproteinase-mediated pathway is important for SARS-CoV-2 infection in addition to the TMPRSS2-mediated pathway and the endosomal pathway. The metalloproteinase-mediated pathway requires both the prior cleavage of spike into two domains and a specific sequence in the second domain, S2, conditions met by SARS-CoV-2 but lacking in the related human coronavirus SARS-CoV. Besides the contribution of metalloproteinases to SARS-CoV-2 infection, inhibition of metalloproteinases was important in preventing cell death, which may cause organ damage. Our study provides new insights into the complex pathogenesis unique to COVID-19 and relevant to the development of effective therapies.
- Published
- 2022
- Full Text
- View/download PDF
9. Development of chimeric receptor activator of nuclear factor-kappa B with glutathione S-transferase in the extracellular domain: Artificial switch in a membrane receptor.
- Author
-
Kawashima K, Hirota-Tsukimachi M, Toma T, Koga R, Iwamaru K, Kanemaru Y, Yanae M, Ahagon A, Nakamura Y, Anraku K, Tateishi H, Gohda J, Inoue JI, Otsuka M, and Fujita M
- Subjects
- Chimera metabolism, Glutathione Transferase genetics, Glutathione Transferase metabolism, Membrane Glycoproteins chemistry, Membrane Glycoproteins metabolism, Osteoclasts metabolism, Receptor Activator of Nuclear Factor-kappa B metabolism, NF-kappa B metabolism, RANK Ligand
- Abstract
Various chimeric receptors have been developed and used for biological experiments. In the present study, we constructed three types of chimeric receptor activator of nuclear factor-kappa B (RANK) with the glutathione S-transferase (GST) protein in the extracellular domain, and stimulated them using newly synthesized chemical trimerizers with three glutathiones. Although this stimulation did not activate these proteins, we unexpectedly found that the chimera named RANK-GST-SC, in which GST replaced a major part of the RANK extracellular domain, activated nuclear factor-kappa B (NF-κB) signaling approximately sixfold more strongly than wild-type RANK without the ligand. The dimerization of extracellular GST is considered to function as a switch outside the cell, and signal transduction then occurs. GST has been widely employed as a tag for protein purification; GST-fusion protein can be conveniently captured by glutathione-conjugated beads and easily purified from impurity. The present study is a pioneering example of the novel utility of GST and provides information for the development of new chemical biology systems., (© 2021 John Wiley & Sons A/S.)
- Published
- 2022
- Full Text
- View/download PDF
10. Identification and characterization of Stathmin 1 as a host factor involved in HIV-1 latency.
- Author
-
Deletsu SD, Kitamura H, Ishida T, Gohda J, Yamaoka S, and Takeuchi H
- Subjects
- HIV Infections genetics, Host-Pathogen Interactions, Humans, RNA Interference, Stathmin genetics, THP-1 Cells, HIV Infections metabolism, HIV-1 physiology, Stathmin metabolism, Virus Latency
- Abstract
Latency remains a barrier to achieving a sterilizing cure to HIV infection. It is thus important to find new host factor(s) to better understand maintenance of HIV latency and be exploited to develop new and more efficient latency reversing agents (LRAs). Here we employed RNA interference screening with a latently HIV-1-infected cell-line to identify Stathmin 1 (STMN1) as a host factor required for maintaining HIV-1 latency. Depletion of STMN1 significantly enhanced HIV-1 expression in a STMN1 depletion-dependent manner and forced expression of exogenous STMN1 suppressed it. We further showed that STMN1 depletion increases HIV-1 proviral transcriptional elongation. Moreover, chromatin immunoprecipitation (ChIP)-qPCR assays revealed STMN1 accumulation on/near the HIV-1 5' LTR region compared to other regions on the HIV-1 provirus, suggesting the possible contribution of STMN1 to HIV-1 transcription. These results suggest that STMN1 is required for the maintenance of HIV-1 latency and implicates STMN1 as a novel therapeutic target to eradicate HIV-1., Competing Interests: Declaration of competing interest No competing interests exists., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
11. N -(4-Hydroxyphenyl) Retinamide Suppresses SARS-CoV-2 Spike Protein-Mediated Cell-Cell Fusion by a Dihydroceramide Δ4-Desaturase 1-Independent Mechanism.
- Author
-
Hayashi Y, Tsuchiya K, Yamamoto M, Nemoto-Sasaki Y, Tanigawa K, Hama K, Ueda Y, Tanikawa T, Gohda J, Maeda K, Inoue JI, and Yamashita A
- Subjects
- Cell Fusion, Cell Membrane genetics, Gene Knockout Techniques, HEK293 Cells, Humans, Membrane Fluidity genetics, Oxidoreductases deficiency, SARS-CoV-2 genetics, Spike Glycoprotein, Coronavirus genetics, Cell Membrane metabolism, Fenretinide pharmacology, Membrane Fluidity drug effects, Oxidoreductases metabolism, SARS-CoV-2 metabolism, Spike Glycoprotein, Coronavirus metabolism
- Abstract
The membrane fusion between the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host cells is essential for the initial step of infection; therefore, the host cell membrane components, including sphingolipids, influence the viral infection. We assessed several inhibitors of the enzymes pertaining to sphingolipid metabolism, against SARS-CoV-2 spike protein (S)-mediated cell-cell fusion and viral infection. N -(4-Hydroxyphenyl) retinamide (4-HPR), an inhibitor of dihydroceramide Δ4-desaturase 1 (DES1), suppressed cell-cell fusion and viral infection. The analysis of sphingolipid levels revealed that the inhibition efficiencies of cell-cell fusion and viral infection in 4-HPR-treated cells were consistent with an increased ratio of saturated sphinganine-based lipids to total sphingolipids. We investigated the relationship of DES1 with the inhibition efficiencies of cell-cell fusion. The changes in the sphingolipid profile induced by 4-HPR were mitigated by the supplementation with exogenous cell-permeative ceramide; however, the reduced cell-cell fusion could not be reversed. The efficiency of cell-cell fusion in DES1 knockout (KO) cells was at a level comparable to that in wild-type (WT) cells; however, the ratio of saturated sphinganine-based lipids to the total sphingolipids was higher in DES1 KO cells than in WT cells. 4-HPR reduced cell membrane fluidity without any significant effects on the expression or localization of angiotensin-converting enzyme 2, the SARS-CoV-2 receptor. Therefore, 4-HPR suppresses SARS-CoV-2 S-mediated membrane fusion through a DES1-independent mechanism, and this decrease in membrane fluidity induced by 4-HPR could be the major cause for the inhibition of SARS-CoV-2 infection. IMPORTANCE Sphingolipids could play an important role in SARS-CoV-2 S-mediated membrane fusion with host cells. We studied the cell-cell fusion using SARS-CoV-2 S-expressing cells and sphingolipid-manipulated target cells, with an inhibitor of the sphingolipid metabolism. 4-HPR (also known as fenretinide) is an inhibitor of DES1, and it exhibits antitumor activity and suppresses cell-cell fusion and viral infection. 4-HPR suppresses membrane fusion through a decrease in membrane fluidity, which could possibly be the cause for the inhibition of SARS-CoV-2 infection. There is accumulating clinical data on the safety of 4-HPR. Therefore, it could be a potential candidate drug against COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
12. Discovery of New Potent anti-MERS CoV Fusion Inhibitors.
- Author
-
Kandeel M, Yamamoto M, Park BK, Al-Taher A, Watanabe A, Gohda J, Kawaguchi Y, Oh-Hashi K, Kwon HJ, and Inoue JI
- Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV), capable of zoonotic transmission, has been associated with emerging viral pneumonia in humans. In this study, a set of highly potent peptides were designed to prevent MERS-CoV fusion through competition with heptad repeat domain 2 (HR2) at its HR1 binding site. We designed eleven peptides with stronger estimated HR1 binding affinities than the wild-type peptide to prevent viral fusion with the cell membrane. Eight peptides showed strong inhibition of spike-mediated MERS-CoV cell-cell fusion with IC50 values in the nanomolar range (0.25-2.3 µM). Peptides #4-6 inhibited 95-98.3% of MERS-CoV plaque formation. Notably, peptide four showed strong inhibition of MERS-CoV plaques formation with EC50 = 0.302 µM. All peptides demonstrated safe profiles without cytotoxicity up to a concentration of 10 μM, and this cellular safety, combined with their anti-MERS-CoV antiviral activity, indicate all peptides can be regarded as potential promising antiviral agents., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Kandeel, Yamamoto, Park, Al-Taher, Watanabe, Gohda, Kawaguchi, Oh-hashi, Kwon and Inoue.)
- Published
- 2021
- Full Text
- View/download PDF
13. Discovery of New Fusion Inhibitor Peptides against SARS-CoV-2 by Targeting the Spike S2 Subunit.
- Author
-
Kandeel M, Yamamoto M, Tani H, Kobayashi A, Gohda J, Kawaguchi Y, Park BK, Kwon HJ, Inoue JI, and Alkattan A
- Abstract
A novel coronavirus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), caused a worldwide pandemic. Our aim in this study is to produce new fusion inhibitors against SARS-CoV-2, which can be the basis for developing new antiviral drugs. The fusion core comprising the heptad repeat domains (HR1 and HR2) of SARS-CoV-2 spike (S) were used to design the peptides. A total of twelve peptides were generated, comprising a short or truncated 24-mer (peptide #1), a long 36-mer peptide (peptide #2), and ten peptide #2 analogs. In contrast to SARS-CoV, SARS-CoV-2 S-mediated cell-cell fusion cannot be inhibited with a minimal length, 24-mer peptide. Peptide #2 demonstrated potent inhibition of SARS-CoV-2 S-mediated cell-cell fusion at 1 µM concentration. Three peptide #2 analogs showed IC50 values in the low micromolar range (4.7-9.8 µM). Peptide #2 inhibited the SARS-CoV-2 pseudovirus assay at IC50=1.49 µM. Given their potent inhibition of viral activity and safety and lack of cytotoxicity, these peptides provide an attractive avenue for the development of new prophylactic and therapeutic agents against SARS-CoV-2.
- Published
- 2021
- Full Text
- View/download PDF
14. TNF receptor-associated factor 6 (TRAF6) plays crucial roles in multiple biological systems through polyubiquitination-mediated NF-κB activation.
- Author
-
Yamamoto M, Gohda J, Akiyama T, and Inoue JI
- Subjects
- Humans, Animals, NF-kappa B metabolism, TNF Receptor-Associated Factor 6 metabolism, Ubiquitination, Signal Transduction
- Abstract
NF-κB was first identified in 1986 as a B cell-specific transcription factor inducing immunoglobulin κ light chain expression. Subsequent studies revealed that NF-κB plays important roles in development, organogenesis, immunity, inflammation, and neurological functions by spatiotemporally regulating cell proliferation, differentiation, and apoptosis in several cell types. Furthermore, studies on the signal pathways that activate NF-κB led to the discovery of TRAF family proteins with E3 ubiquitin ligase activity, which function downstream of the receptor. This discovery led to the proposal of an entirely new signaling mechanism concept, wherein K63-ubiquitin chains act as a scaffold for the signaling complex to activate downstream kinases. This concept has revolutionized ubiquitin studies by revealing the importance of the nonproteolytic functions of ubiquitin not only in NF-κB signaling but also in a variety of other biological systems. TRAF6 is the most diverged among the TRAF family proteins, and our studies uncovered its notable physiological and pathological functions.
- Published
- 2021
- Full Text
- View/download PDF
15. The Anticoagulant Nafamostat Potently Inhibits SARS-CoV-2 S Protein-Mediated Fusion in a Cell Fusion Assay System and Viral Infection In Vitro in a Cell-Type-Dependent Manner.
- Author
-
Yamamoto M, Kiso M, Sakai-Tagawa Y, Iwatsuki-Horimoto K, Imai M, Takeda M, Kinoshita N, Ohmagari N, Gohda J, Semba K, Matsuda Z, Kawaguchi Y, Kawaoka Y, and Inoue JI
- Subjects
- Angiotensin-Converting Enzyme 2, Angiotensin-Converting Enzyme Inhibitors pharmacology, Animals, Benzamidines, Betacoronavirus metabolism, COVID-19, Cell Line, Chlorocebus aethiops, Coronavirus Infections virology, Esters, Gabexate analogs & derivatives, Gabexate pharmacology, HEK293 Cells, Humans, Pandemics, Peptidyl-Dipeptidase A metabolism, Pneumonia, Viral virology, SARS-CoV-2, Serine Endopeptidases metabolism, Spike Glycoprotein, Coronavirus metabolism, Vero Cells, Anticoagulants pharmacology, Betacoronavirus drug effects, Coronavirus Infections drug therapy, Guanidines pharmacology, Pneumonia, Viral drug therapy, Spike Glycoprotein, Coronavirus antagonists & inhibitors, Virus Internalization drug effects
- Abstract
Although infection by SARS-CoV-2, the causative agent of coronavirus pneumonia disease (COVID-19), is spreading rapidly worldwide, no drug has been shown to be sufficiently effective for treating COVID-19. We previously found that nafamostat mesylate, an existing drug used for disseminated intravascular coagulation (DIC), effectively blocked Middle East respiratory syndrome coronavirus (MERS-CoV) S protein-mediated cell fusion by targeting transmembrane serine protease 2 (TMPRSS2), and inhibited MERS-CoV infection of human lung epithelium-derived Calu-3 cells. Here we established a quantitative fusion assay dependent on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) S protein, angiotensin I converting enzyme 2 (ACE2) and TMPRSS2, and found that nafamostat mesylate potently inhibited the fusion while camostat mesylate was about 10-fold less active. Furthermore, nafamostat mesylate blocked SARS-CoV-2 infection of Calu-3 cells with an effective concentration (EC)
50 around 10 nM, which is below its average blood concentration after intravenous administration through continuous infusion. On the other hand, a significantly higher dose (EC50 around 30 mM) was required for VeroE6/TMPRSS2 cells, where the TMPRSS2-independent but cathepsin-dependent endosomal infection pathway likely predominates. Together, our study shows that nafamostat mesylate potently inhibits SARS-CoV-2 S protein-mediated fusion in a cell fusion assay system and also inhibits SARS-CoV-2 infection in vitro in a cell-type-dependent manner. These findings, together with accumulated clinical data regarding nafamostat's safety, make it a likely candidate drug to treat COVID-19.- Published
- 2020
- Full Text
- View/download PDF
16. Structural analysis of TIFA: Insight into TIFA-dependent signal transduction in innate immunity.
- Author
-
Nakamura T, Hashikawa C, Okabe K, Yokote Y, Chirifu M, Toma-Fukai S, Nakamura N, Matsuo M, Kamikariya M, Okamoto Y, Gohda J, Akiyama T, Semba K, Ikemizu S, Otsuka M, Inoue JI, and Yamagata Y
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Humans, Intracellular Signaling Peptides and Proteins physiology, Mice, NF-kappa B metabolism, Pathogen-Associated Molecular Pattern Molecules immunology, Pathogen-Associated Molecular Pattern Molecules metabolism, Phosphorylation, Protein Kinases metabolism, Signal Transduction physiology, TNF Receptor-Associated Factor 6 metabolism, Tumor Necrosis Factor Receptor-Associated Peptides and Proteins metabolism, Ubiquitination, Immunity, Innate immunology, Intracellular Signaling Peptides and Proteins metabolism, Tumor Necrosis Factor Receptor-Associated Peptides and Proteins ultrastructure
- Abstract
TRAF-interacting protein with a forkhead-associated (FHA) domain (TIFA), originally identified as an adaptor protein of TRAF6, has recently been shown to be involved in innate immunity, induced by a pathogen-associated molecular pattern (PAMP). ADP-β-D-manno-heptose, a newly identified PAMP, binds to alpha-kinase 1 (ALPK1) and activates its kinase activity to phosphorylate TIFA. Phosphorylation triggers TIFA oligomerisation and formation of a subsequent TIFA-TRAF6 oligomeric complex for ubiquitination of TRAF6, eventually leading to NF-κB activation. However, the structural basis of TIFA-dependent TRAF6 signalling, especially oligomer formation of the TIFA-TRAF6 complex remains unknown. In the present study, we determined the crystal structures of mouse TIFA and two TIFA mutants-Thr9 mutated to either Asp or Glu to mimic the phosphorylation state-to obtain the structural information for oligomer formation of the TIFA-TRAF6 complex. Crystal structures show the dimer formation of mouse TIFA to be similar to that of human TIFA, which was previously reported. This dimeric structure is consistent with the solution structure obtained from small angle X-ray scattering analysis. In addition to the structural analysis, we examined the molecular assembly of TIFA and the TIFA-TRAF6 complex by size-exclusion chromatography, and suggested a model for the TIFA-TRAF6 signalling complex.
- Published
- 2020
- Full Text
- View/download PDF
17. BI-2536 and BI-6727, dual Polo-like kinase/bromodomain inhibitors, effectively reactivate latent HIV-1.
- Author
-
Gohda J, Suzuki K, Liu K, Xie X, Takeuchi H, Inoue JI, Kawaguchi Y, and Ishida T
- Subjects
- Cell Line, Tumor, Drug Combinations, HIV-1 genetics, HIV-1 growth & development, Histone Deacetylase Inhibitors pharmacology, Host-Pathogen Interactions drug effects, Host-Pathogen Interactions genetics, Humans, Inhibitory Concentration 50, Leukocytes, Mononuclear drug effects, Leukocytes, Mononuclear metabolism, Leukocytes, Mononuclear virology, Lymphocytes drug effects, Lymphocytes metabolism, Lymphocytes virology, Monocytes drug effects, Monocytes metabolism, Monocytes virology, Phorbol Esters pharmacology, Primary Cell Culture, Protein Domains, Protein Kinase Inhibitors pharmacology, RNA, Messenger agonists, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Viral agonists, RNA, Viral metabolism, THP-1 Cells, Transcription, Genetic drug effects, Virus Activation genetics, Virus Latency genetics, Vorinostat pharmacology, Gene Expression Regulation, Viral drug effects, HIV-1 drug effects, Pteridines pharmacology, RNA, Viral genetics, Virus Activation drug effects, Virus Latency drug effects
- Abstract
HIV-1 latent reservoirs harbouring silenced but replication-competent proviruses are a major obstacle against viral eradication in infected patients. The "shock and kill" strategy aims to reactivate latent provirus with latency reversing agents (LRAs) in the presence of antiretroviral drugs, necessitating the development of effective and efficient LRAs. We screened a chemical library for potential LRAs and identified two dual Polo-like kinase (PLK)/bromodomain inhibitors, BI-2536 and BI-6727 (volasertib), which are currently undergoing clinical trials against various cancers. BI-2536 and BI-6727 significantly reactivated silenced HIV-1 provirus at both the mRNA and protein level in two latently infected model cell lines (ACH2 and U1). BI-2536 dramatically reactivated transcription of latent HIV-1 provirus in peripheral blood mononuclear cells derived from infected patients. Long terminal repeat activation by the inhibitors was associated with bromodomain rather than PLK inhibition. We also found that BI-2536 synergistically activates the latent provirus in combination with SAHA, a histone deacetylase inhibitor, or the non-tumour-promoting phorbol ester prostratin. Our findings strongly suggest that BI-2536 and BI-6727 are potent LRAs for the "shock and kill" HIV-1 eradication strategy.
- Published
- 2018
- Full Text
- View/download PDF
18. miR-146a - Traf6 regulatory axis controls autoimmunity and myelopoiesis, but is dispensable for hematopoietic stem cell homeostasis and tumor suppression.
- Author
-
Magilnick N, Reyes EY, Wang WL, Vonderfecht SL, Gohda J, Inoue JI, and Boldin MP
- Subjects
- Animals, Female, Gene Expression Regulation, Hematopoietic Stem Cells immunology, Homeostasis, Humans, Inflammation genetics, Inflammation physiopathology, Male, Mice, MicroRNAs genetics, Myeloid Cells cytology, Myeloid Cells immunology, Neoplasms genetics, Neoplasms physiopathology, TNF Receptor-Associated Factor 6 genetics, Autoimmunity, Hematopoietic Stem Cells cytology, Inflammation immunology, MicroRNAs immunology, Myelopoiesis, Neoplasms immunology, TNF Receptor-Associated Factor 6 immunology
- Abstract
microRNA-146a ( miR-146a ) has been previously implicated as an essential molecular brake, preventing immune overreaction and malignant transformation by attenuating NF-κB signaling, putatively via repression of the Traf6 and Irak1 genes. The exact contribution of miR-146a -mediated silencing of these genes to the control of immune activation is currently unknown. Therefore, we defined the role of the miR-146a - Traf6 signaling axis in the regulation of immune homeostasis using a genetic epistasis analysis in miR-146a
-/- mice. We have uncovered a surprising separation of functions at the level of miR-146a targets. Lowering the Traf6 gene dose and consequent attenuation of NF-κB activation rescued several significant miR-146a-/- phenotypes, such as splenomegaly, aberrant myeloproliferation, and excessive inflammatory responses. In contrast, decreasing Traf6 expression had no effect on the development of the progressive bone marrow failure phenotype, as well as lymphomagenesis in miR-146a-/- mice, indicating that miR-146a controls these biological processes through different molecular mechanisms., Competing Interests: The authors declare no conflict of interest.- Published
- 2017
- Full Text
- View/download PDF
19. Basal autophagy prevents autoactivation or enhancement of inflammatory signals by targeting monomeric MyD88.
- Author
-
Into T, Horie T, Inomata M, Gohda J, Inoue JI, Murakami Y, and Niida S
- Subjects
- Animals, Cells, Cultured, Female, Inflammasomes metabolism, Intracellular Signaling Peptides and Proteins, Male, Mice, Myeloid Differentiation Factor 88 chemistry, Myeloid Differentiation Factor 88 genetics, Signal Transduction, TNF Receptor-Associated Factor 6 genetics, Autophagy, Lysosomes metabolism, Myeloid Differentiation Factor 88 metabolism, TNF Receptor-Associated Factor 6 metabolism
- Abstract
Autophagy, the processes of delivery of intracellular components to lysosomes, regulates induction of inflammation. Inducible macroautophagy degrades inflammasomes and dysfunctional mitochondria to downregulate inflammatory signals. Nonetheless, the effects of constitutive basal autophagy on inflammatory signals are largely unknown. Here, we report a previously unknown effect of basal autophagy. Lysosomal inhibition induced weak inflammatory signals in the absence of a cellular stimulus and in the presence of a nutrient supply, and their induction was impaired by MyD88 deficiency. During lysosomal inhibition, MyD88 was accumulated, and overabundant MyD88 autoactivated downstream signaling or enhanced TLR/IL-1R-mediated signaling. MyD88 is probably degraded via basal microautophagy because macroautophagy inhibitors, ATG5 deficiency, and an activator of chaperone-mediated autophagy did not affect MyD88. Analysis using a chimeric protein whose monomerization/dimerization can be switched revealed that monomeric MyD88 is susceptible to degradation. Immunoprecipitation of monomeric MyD88 revealed its interaction with TRAF6. In TRAF6-deficient cells, degradation of basal MyD88 was enhanced, suggesting that TRAF6 participates in protection from basal autophagy. Thus, basal autophagy lowers monomeric MyD88 expression, and thereby autoactivation of inflammatory signals is prevented. Given that impairment of lysosomes occurs in various settings, our results provide novel insights into the etiology of inflammatory signals that affect consequences of inflammation.
- Published
- 2017
- Full Text
- View/download PDF
20. HTLV-1 Tax Induces Formation of the Active Macromolecular IKK Complex by Generating Lys63- and Met1-Linked Hybrid Polyubiquitin Chains.
- Author
-
Shibata Y, Tokunaga F, Goto E, Komatsu G, Gohda J, Saeki Y, Tanaka K, Takahashi H, Sawasaki T, Inoue S, Oshiumi H, Seya T, Nakano H, Tanaka Y, Iwai K, and Inoue JI
- Subjects
- Electrophoresis, Polyacrylamide Gel, HEK293 Cells, Human T-lymphotropic virus 1 pathogenicity, Humans, Immunoblotting, Immunoprecipitation, Jurkat Cells, Real-Time Polymerase Chain Reaction, Signal Transduction physiology, Transfection, Enzyme Activation physiology, Gene Products, tax metabolism, HTLV-I Infections metabolism, I-kappa B Kinase metabolism, NF-kappa B metabolism
- Abstract
The Tax protein of human T-cell leukemia virus type 1 (HTLV-1) is crucial for the development of adult T-cell leukemia (ATL), a highly malignant CD4+ T cell neoplasm. Among the multiple aberrant Tax-induced effects on cellular processes, persistent activation of transcription factor NF-κB, which is activated only transiently upon physiological stimulation, is essential for leukemogenesis. We and others have shown that Tax induces activation of the IκB kinase (IKK) complex, which is a critical step in NF-κB activation, by generating Lys63-linked polyubiquitin chains. However, the molecular mechanism underlying Tax-induced IKK activation is controversial and not fully understood. Here, we demonstrate that Tax recruits linear (Met1-linked) ubiquitin chain assembly complex (LUBAC) to the IKK complex and that Tax fails to induce IKK activation in cells that lack LUBAC activity. Mass spectrometric analyses revealed that both Lys63-linked and Met1-linked polyubiquitin chains are associated with the IKK complex. Furthermore, treatment of the IKK-associated polyubiquitin chains with Met1-linked-chain-specific deubiquitinase (OTULIN) resulted in the reduction of high molecular weight polyubiquitin chains and the generation of short Lys63-linked ubiquitin chains, indicating that Tax can induce the generation of Lys63- and Met1-linked hybrid polyubiquitin chains. We also demonstrate that Tax induces formation of the active macromolecular IKK complex and that the blocking of Tax-induced polyubiquitin chain synthesis inhibited formation of the macromolecular complex. Taken together, these results lead us to propose a novel model in which the hybrid-chain-dependent oligomerization of the IKK complex triggered by Tax leads to trans-autophosphorylation-mediated IKK activation., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2017
- Full Text
- View/download PDF
21. Visualization of RelB expression and activation at the single-cell level during dendritic cell maturation in Relb-Venus knock-in mice.
- Author
-
Seki T, Yamamoto M, Taguchi Y, Miyauchi M, Akiyama N, Yamaguchi N, Gohda J, Akiyama T, and Inoue J
- Subjects
- Animals, B-Lymphocytes metabolism, B7-2 Antigen metabolism, Bacterial Proteins genetics, Bacterial Proteins metabolism, Cell Differentiation genetics, Cells, Cultured, Dendritic Cells cytology, Female, Gene Expression Regulation, Gene Knock-In Techniques, Genes, MHC Class II, Genes, Reporter, Luminescent Proteins genetics, Luminescent Proteins metabolism, Male, Mice, Mice, Inbred C57BL, Signal Transduction, Spleen cytology, Thymus Gland cytology, Transcription Factor RelB genetics, Dendritic Cells immunology, Single-Cell Analysis, Transcription Factor RelB metabolism
- Abstract
RelB is activated by the non-canonical NF-κB pathway, which is crucial for immunity by establishing lymphoid organogenesis and B-cell and dendritic cell (DC) maturation. To elucidate the mechanism of the RelB-mediated immune cell maturation, a precise understanding of the relationship between cell maturation and RelB expression and activation at the single-cell level is required. Therefore, we generated knock-in mice expressing a fusion protein between RelB and fluorescent protein (RelB-Venus) from the Relb locus. The Relb(Venus/Venus) mice developed without any abnormalities observed in the Relb(-/-) mice, allowing us to monitor RelB-Venus expression and nuclear localization as RelB expression and activation. Relb(Venus/Venus) DC analyses revealed that DCs consist of RelB(-), RelB(low) and RelB(high) populations. The RelB(high) population, which included mature DCs with projections, displayed RelB nuclear localization, whereas RelB in the RelB(low) population was in the cytoplasm. Although both the RelB(low) and RelB(-) populations barely showed projections, MHC II and co-stimulatory molecule expression were higher in the RelB(low) than in the RelB(-) splenic conventional DCs. Taken together, our results identify the RelB(low) population as a possible novel intermediate maturation stage of cDCs and the Relb(Venus/Venus) mice as a useful tool to analyse the dynamic regulation of the non-canonical NF-κB pathway., (© The Authors 2015. Published by Oxford University Press on behalf of the Japanese Biochemical Society. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
22. Loss of Tifab, a del(5q) MDS gene, alters hematopoiesis through derepression of Toll-like receptor-TRAF6 signaling.
- Author
-
Varney ME, Niederkorn M, Konno H, Matsumura T, Gohda J, Yoshida N, Akiyama T, Christie S, Fang J, Miller D, Jerez A, Karsan A, Maciejewski JP, Meetei RA, Inoue J, and Starczynowski DT
- Subjects
- Animals, Apoptosis, Bone Marrow Transplantation, Cell Differentiation, Chromosomes, Human, Pair 5, Humans, Intracellular Signaling Peptides and Proteins, Male, Mice, Mice, Inbred C57BL, MicroRNAs physiology, NF-kappa B antagonists & inhibitors, NF-kappa B physiology, Proteins genetics, Hematopoiesis, Proteins physiology, Signal Transduction physiology, TNF Receptor-Associated Factor 6 physiology, Toll-Like Receptors physiology
- Abstract
TRAF-interacting protein with forkhead-associated domain B (TIFAB) is a haploinsufficient gene in del(5q) myelodysplastic syndrome (MDS). Deletion of Tifab results in progressive bone marrow (BM) and blood defects, including skewed hematopoietic stem/progenitor cell (HSPC) proportions and altered myeloid differentiation. A subset of mice transplanted with Tifab knockout (KO) HSPCs develop a BM failure with neutrophil dysplasia and cytopenia. In competitive transplants, Tifab KO HSPCs are out-competed by wild-type (WT) cells, suggesting a cell-intrinsic defect. Gene expression analysis of Tifab KO HSPCs identified dysregulation of immune-related signatures, and hypersensitivity to TLR4 stimulation. TIFAB forms a complex with TRAF6, a mediator of immune signaling, and reduces TRAF6 protein stability by a lysosome-dependent mechanism. In contrast, TIFAB loss increases TRAF6 protein and the dynamic range of TLR4 signaling, contributing to ineffective hematopoiesis. Moreover, combined deletion of TIFAB and miR-146a, two genes associated with del(5q) MDS/AML, results in a cooperative increase in TRAF6 expression and hematopoietic dysfunction. Re-expression of TIFAB in del(5q) MDS/AML cells results in attenuated TLR4 signaling and reduced viability. These findings underscore the importance of efficient regulation of innate immune/TRAF6 signaling within HSPCs by TIFAB, and its cooperation with miR-146a as it relates to the pathogenesis of hematopoietic malignancies, such as del(5q) MDS/AML., (© 2015 Varney et al.)
- Published
- 2015
- Full Text
- View/download PDF
23. A RANKL mutant used as an inter-species vaccine for efficient immunotherapy of osteoporosis.
- Author
-
Liu C, Zhao Y, He W, Wang W, Chen Y, Zhang S, Ma Y, Gohda J, Ishida T, Walter TS, Owens RJ, Stuart DI, Ren J, and Gao B
- Subjects
- Animals, Antibodies immunology, Autoantigens immunology, Autoantigens pharmacology, Bone Resorption genetics, Bone Resorption metabolism, Cell Differentiation drug effects, Disease Models, Animal, Humans, Immunization, Mice, Models, Molecular, Osteoclasts cytology, Osteoclasts immunology, Osteoclasts metabolism, Osteoporosis diagnosis, Osteoporosis therapy, Ovariectomy, Protein Binding, Protein Conformation, RANK Ligand chemistry, RANK Ligand metabolism, Rats, Receptor Activator of Nuclear Factor-kappa B chemistry, Receptor Activator of Nuclear Factor-kappa B metabolism, X-Ray Microtomography, Immunotherapy, Osteoporosis genetics, Osteoporosis immunology, RANK Ligand genetics, RANK Ligand immunology, Vaccines
- Abstract
Anti-cytokine therapeutic antibodies have been demonstrated to be effective in the treatment of several auto-immune disorders. However, The problems in antibody manufacture and the immunogenicity caused by multiple doses of antibodies inspire people to use auto-cytokine as immunogen to induce anti-cytokine antibodies. Nevertheless, the tolerance for inducing immune response against self-antigen has hindered the wide application of the strategy. To overcome the tolerance, here we proposed a strategy using the inter-species cytokine as immunogen for active immunization (TISCAI) to induce anti-cytokine antibody. As a proof of concept, an inter-species cytokine RANKL was successfully used as immunogen to induce anti-RANKL immune response. Furthermore, to prevent undesirable side-effects, the human RANKL was mutated based on the crystal structure of the complex of human RANKL and its rodent counterpart receptor RANK. We found, the antibodies produced blocked the osteoclast development in vitro and osteoporosis in OVX rat models. The results demonstrated this strategy adopted is very useful for general anti-cytokine immunotherapy for different diseases settings.
- Published
- 2015
- Full Text
- View/download PDF
24. Emergence of Lamivudine-Resistant HBV during Antiretroviral Therapy Including Lamivudine for Patients Coinfected with HIV and HBV in China.
- Author
-
Gu L, Han Y, Li Y, Zhu T, Song X, Huang Y, Yang F, Guan S, Xie J, Gohda J, Hosoya N, Kawana-Tachikawa A, Liu W, Gao GF, Iwamoto A, Li T, and Ishida T
- Subjects
- Adolescent, Adult, Anti-Retroviral Agents pharmacology, China epidemiology, Coinfection epidemiology, Female, HIV Infections epidemiology, HIV-1 drug effects, HIV-1 physiology, Hepatitis B epidemiology, Hepatitis B virus physiology, Humans, Lamivudine pharmacology, Male, Middle Aged, Virus Replication drug effects, Young Adult, Anti-Retroviral Agents therapeutic use, Coinfection drug therapy, Drug Resistance, Viral, HIV Infections drug therapy, Hepatitis B drug therapy, Hepatitis B virus drug effects, Lamivudine therapeutic use
- Abstract
In China, HIV-1-infected patients typically receive antiretroviral therapy (ART) that includes lamivudine (3TC) as a reverse-transcriptase inhibitor (RTI) (ART-3TC). Previous studies from certain developed countries have shown that, in ART-3TC, 3TC-resistant HBV progressively emerges at an annual rate of 15-20% in patients coinfected with HIV-1 and HBV. This scenario in China warrants investigation because >10% of all HIV-infected patients in China are HBV carriers. We measured the occurrence of 3TC-resistant HBV during ART-3TC for HIV-HBV coinfection and also tested the effect of tenofovir disoproxil fumarate (TDF) used as an additional RTI (ART-3TC/TDF) in a cohort study in China. We obtained 200 plasma samples collected from 50 Chinese patients coinfected with HIV-1 and HBV (positive for hepatitis B surface antigen) and examined them for the prevalence of 3TC-resistant HBV by directly sequencing PCR products that covered the HBV reverse-transcriptase gene. We divided the patients into ART-3TC and ART-3TC/TDF groups and compared the efficacy of treatment and incidence of drug-resistance mutation between the groups. HIV RNA and HBV DNA loads drastically decreased in both ART-3TC and ART-3TC/TDF groups. In the ART-3TC group, HBV breakthrough or insufficient suppression of HBV DNA loads was observed in 20% (10/50) of the patients after 96-week treatment, and 8 of these patients harbored 3TC-resistant mutants. By contrast, neither HBV breakthrough nor treatment failure was recorded in the ART-3TC/TDF group. All of the 3TC-resistant HBV mutants emerged from the cases in which HBV DNA loads were high at baseline. Our results clearly demonstrated that ART-3TC is associated with the emergence of 3TC-resistant HBV in patients coinfected with HIV-1 and HBV and that ART-3TC/TDF reduces HBV DNA loads to an undetectable level. These findings support the use of TDF-based treatment regimens for patients coinfected with HIV-1 and HBV.
- Published
- 2015
- Full Text
- View/download PDF
25. HIV-1 replicates in human osteoclasts and enhances their differentiation in vitro.
- Author
-
Gohda J, Ma Y, Huang Y, Zhang Y, Gu L, Han Y, Li T, Gao B, Gao GF, Inoue J, Iwamoto A, and Ishida T
- Subjects
- CD4 Antigens analysis, Cells, Cultured, Humans, Lipopolysaccharide Receptors analysis, Osteoclasts chemistry, Receptors, CCR5 analysis, Receptors, CXCR4 analysis, Cell Differentiation, HIV-1 physiology, Osteoclasts physiology, Osteoclasts virology, Virus Replication
- Abstract
Background: HIV-1 infected patients frequently have osteolytic bone disease, which is caused by the dysregulation of the bone remodeling system that involves the interaction between osteoblasts and osteoclasts, but the relationship between osteolytic disease and HIV-1 infection remains unclear. In this study we tested whether HIV-1 infection of osteoclasts affects their differentiation., Results: We prepared human osteoclasts from CD14+ monocytes and examined them for their susceptibility to HIV-1. Furthermore, we investigated the effect of HIV-1 infection on osteoclast differentiation. CD14-derived osteoclasts were shown to express CD4, CCR5, and CXCR4 each at the similar level to that shown with macrophages. R5-tropic HIV-1 and X4-tropic HIV-1 were found to infect CD14-derived osteoclasts and replicate in them. Furthermore, HIV-1 infection induced formation of larger osteoclastst, enhanced the expression of mRNAs for three osteoclast specific marker molecules (tartrate-resistant acid phosphatase, cathepsin K, and the calcitonin receptor), and up-regulated osteoclast bone resorption activity., Conclusions: Our results suggest that osteoclasts serve as a novel target for HIV-1 infection, which may enhance the osteoclast differentiation contributing to the development of osteolytic disease in HIV-1-infected patients.
- Published
- 2015
- Full Text
- View/download PDF
26. cIAP1/2 negatively regulate RANKL-induced osteoclastogenesis through the inhibition of NFATc1 expression.
- Author
-
Yamaguchi N, Yokota M, Taguchi Y, Gohda J, and Inoue J
- Subjects
- Animals, Cell Line, Gene Expression Regulation, Inhibitor of Apoptosis Proteins antagonists & inhibitors, MAP Kinase Signaling System, Mice, Mice, Inbred C57BL, Monocytes cytology, Monocytes metabolism, NF-kappa B metabolism, NFATC Transcription Factors genetics, Osteoclasts cytology, RNA, Messenger biosynthesis, RNA, Small Interfering, Cell Differentiation, Inhibitor of Apoptosis Proteins metabolism, NFATC Transcription Factors metabolism, Osteoclasts metabolism, RANK Ligand metabolism
- Abstract
Receptor activator of nuclear factor κB (RANK) is a member of the tumor necrosis factor receptor superfamily (TNFRSF) and triggers osteoclastogenesis by inducing the expression of NFATc1 through the activation of the NF-κB and MAPK pathways. Cellular inhibitors of apoptosis proteins 1 and 2 (cIAP1/2), which are ubiquitin E3 ligases, are involved in the activation of the NF-κB and MAPK pathways by various members of the TNFRSF. However, the involvement of cIAP1/2 in RANK signaling has remained largely unknown. In this study, we reveal the involvement of cIAP1/2 in RANK ligand (RANKL)-induced osteoclastogenesis. The over-expression of cIAP1 or cIAP2 in the mouse monocytic cell line Raw264.7 resulted in the significant suppression of RANKL-induced NFATc1 mRNA expression and osteoclastogenesis, whereas the activation of the NF-κB and MAPK pathways was barely changed by these over-expressions. The depletion of endogenous cIAP1/2 by their specific inhibitor MV1 or their siRNA-mediated knockdown resulted in enhanced RANKL-induced NFATc1 expression and osteoclastogenesis without affecting the activation of the NF-κB and MAPK pathways. In combination, these results indicate that cIAP1/2 negatively regulate osteoclastogenesis by inhibiting NFATc1 mRNA expression in a manner that is distinct from the previously identified functions of cIAP1/2., (© 2012 The Authors Genes to Cells © 2012 by the Molecular Biology Society of Japan and Wiley Publishing Asia Pty Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
27. Identification and characterization of anti-osteoclastogenic peptides derived from the cytoplasmic tail of receptor activator of nuclear factor kappa B.
- Author
-
Taguchi Y, Kiga Y, Gohda J, and Inoue J
- Subjects
- Adaptor Proteins, Signal Transducing, Amino Acid Sequence, Animals, Bone Marrow Cells metabolism, C-Peptide genetics, C-Peptide metabolism, Cell Fusion, Cytoplasm genetics, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Peptides genetics, Phosphoproteins genetics, Phosphoproteins metabolism, Protein Binding genetics, Protein Structure, Tertiary genetics, Proto-Oncogene Proteins c-vav genetics, Proto-Oncogene Proteins c-vav metabolism, Receptor Activator of Nuclear Factor-kappa B genetics, Signal Transduction genetics, TNF Receptor-Associated Factor 6 genetics, TNF Receptor-Associated Factor 6 metabolism, Cytoplasm metabolism, Osteoclasts metabolism, Peptides metabolism, Receptor Activator of Nuclear Factor-kappa B metabolism
- Abstract
Pathological bone resorption by osteoclasts is primarily treated with bisphosphonates. Because the administration of bisphosphonates is associated with a risk for multiple adverse symptoms, a precise understanding of the mechanisms underlying osteoclastogenesis is required to develop drugs with minimal side-effects. Osteoclastogenesis depends on receptor activator of nuclear factor kappa B (RANK) signaling mediated by TRAF6. We previously identified a highly conserved domain in the cytoplasmic tail of RANK (HCR), which did not share any significant homology with other proteins and was essential for osteoclastogenesis. HCR acts as a platform for the formation of Gab2- and Vav3-containing signal complexes, and ectopic expression of the HCR peptide inhibits osteoclastogenesis. Here, we uncover the mechanisms of HCR peptide-mediated inhibition of osteoclastogenesis. Expression of either the amino- or carboxyl-terminal half of the HCR peptide (N- or C-peptide) independently inhibited RANK signaling prior to cell-cell fusion. In contrast, expression of the GY-peptide, which is a part of the C-peptide, did not significantly affect prefusion RANK signaling, but did inhibit cell-cell fusion to prevent formation of multinucleated mature osteoclasts. Moreover, Gab2, which is involved in RANK signaling by binding TRAF6, bound the C-peptide but not the N-peptide, suggesting that the C- and the N-peptides sequester TRAF6 in a Gab2-dependent and Gab2-independent manner, respectively. In contrast, the GY-peptide did not bind Gab2 but could bind Vav3, which mediates signaling for cell-cell fusion. Collectively, we propose that the HCR peptide inhibits osteoclastogenesis through two modes of action-inhibition of (1) prefusion RANK signaling and (2) cell-cell fusion by blocking TRAF6- and Vav3-mediated signaling, respectively.
- Published
- 2012
- Full Text
- View/download PDF
28. The Shigella flexneri effector OspI deamidates UBC13 to dampen the inflammatory response.
- Author
-
Sanada T, Kim M, Mimuro H, Suzuki M, Ogawa M, Oyama A, Ashida H, Kobayashi T, Koyama T, Nagai S, Shibata Y, Gohda J, Inoue J, Mizushima T, and Sasakawa C
- Subjects
- Amidohydrolases genetics, Amino Acid Sequence, Animals, Aspartic Acid metabolism, B-Cell CLL-Lymphoma 10 Protein, Biocatalysis, Caspases metabolism, Catalytic Domain genetics, Crystallography, X-Ray, Cysteine metabolism, DNA Mutational Analysis, Diglycerides antagonists & inhibitors, Diglycerides metabolism, Dysentery, Bacillary microbiology, Glutamic Acid metabolism, Glutamine metabolism, HEK293 Cells, HeLa Cells, Histidine metabolism, Humans, Immunity, Innate, Inflammation enzymology, Mice, Models, Molecular, Molecular Sequence Data, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, NF-kappa B metabolism, Neoplasm Proteins metabolism, Shigella flexneri genetics, Shigella flexneri pathogenicity, TNF Receptor-Associated Factor 6 deficiency, TNF Receptor-Associated Factor 6 genetics, TNF Receptor-Associated Factor 6 metabolism, Ubiquitin-Conjugating Enzymes chemistry, Ubiquitin-Conjugating Enzymes genetics, Virulence Factors metabolism, Adaptor Proteins, Signal Transducing metabolism, Amidohydrolases chemistry, Amidohydrolases metabolism, Inflammation immunology, Inflammation metabolism, Shigella flexneri enzymology, Shigella flexneri immunology, Ubiquitin-Conjugating Enzymes metabolism
- Abstract
Many bacterial pathogens can enter various host cells and then survive intracellularly, transiently evade humoral immunity, and further disseminate to other cells and tissues. When bacteria enter host cells and replicate intracellularly, the host cells sense the invading bacteria as damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) by way of various pattern recognition receptors. As a result, the host cells induce alarm signals that activate the innate immune system. Therefore, bacteria must modulate host inflammatory signalling and dampen these alarm signals. How pathogens do this after invading epithelial cells remains unclear, however. Here we show that OspI, a Shigella flexneri effector encoded by ORF169b on the large plasmid and delivered by the type ΙΙΙ secretion system, dampens acute inflammatory responses during bacterial invasion by suppressing the tumour-necrosis factor (TNF)-receptor-associated factor 6 (TRAF6)-mediated signalling pathway. OspI is a glutamine deamidase that selectively deamidates the glutamine residue at position 100 in UBC13 to a glutamic acid residue. Consequently, the E2 ubiquitin-conjugating activity required for TRAF6 activation is inhibited, allowing S. flexneri OspI to modulate the diacylglycerol-CBM (CARD-BCL10-MALT1) complex-TRAF6-nuclear-factor-κB signalling pathway. We determined the 2.0 Å crystal structure of OspI, which contains a putative cysteine-histidine-aspartic acid catalytic triad. A mutational analysis showed this catalytic triad to be essential for the deamidation of UBC13. Our results suggest that S. flexneri inhibits acute inflammatory responses in the initial stage of infection by targeting the UBC13-TRAF6 complex.
- Published
- 2012
- Full Text
- View/download PDF
29. p47 negatively regulates IKK activation by inducing the lysosomal degradation of polyubiquitinated NEMO.
- Author
-
Shibata Y, Oyama M, Kozuka-Hata H, Han X, Tanaka Y, Gohda J, and Inoue J
- Subjects
- Adenosine Triphosphatases genetics, Adenosine Triphosphatases metabolism, Cell Line, Electrophoretic Mobility Shift Assay, HeLa Cells, Humans, I-kappa B Kinase genetics, Immunoprecipitation, Jurkat Cells metabolism, NF-kappa B genetics, NF-kappa B metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Protein Binding, Real-Time Polymerase Chain Reaction, Sf9 Cells, Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins genetics, I-kappa B Kinase metabolism, Lysosomes metabolism, Polyubiquitin metabolism, Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins metabolism
- Abstract
The persistent or excess activation of NF-κB causes various inflammatory and autoimmune diseases, but the molecular mechanisms that negatively regulate NF-κB activation are not fully understood. Here we show that p47, an essential factor for Golgi membrane fusion, associates with the NEMO subunit of the IκB kinase (IKK) complex upon TNF-α or IL-1 stimulation, and inhibits IKK activation. p47 binds to Lys63-linked and linear polyubiquitin chains, which are conjugated to NEMO upon such stimulation. The binding of p47 to polyubiquitinated NEMO triggers the lysosomal degradation of NEMO, thereby inhibiting IKK activation. The silencing of p47 results in enhanced TNF-α- or IL-1-induced IKK activation, and an increased expression of genes encoding inflammatory mediators. Taken together, our results suggest that p47 is critical for negatively regulating stimulation-induced IKK activation in a manner that is mechanistically distinct from the previously characterized negative regulators, such as A20 and CYLD.
- Published
- 2012
- Full Text
- View/download PDF
30. Activation of the IκB kinase complex by HTLV-1 Tax requires cytosolic factors involved in Tax-induced polyubiquitination.
- Author
-
Shibata Y, Tanaka Y, Gohda J, and Inoue J
- Subjects
- Baculoviridae genetics, Baculoviridae metabolism, Cytosol metabolism, Enzyme Activation, Gene Products, tax genetics, Gene Products, tax pharmacology, Human T-lymphotropic virus 1 metabolism, Humans, I-kappa B Kinase antagonists & inhibitors, I-kappa B Kinase genetics, Imidazoles pharmacology, Immunoprecipitation, Jurkat Cells, MAP Kinase Kinase Kinase 1 genetics, MAP Kinase Kinase Kinase 1 metabolism, MAP Kinase Kinase Kinase 1 pharmacology, Nitriles pharmacology, Phosphorylation, Plasmids genetics, Plasmids metabolism, Protein Interaction Mapping, Quinoxalines pharmacology, Recombinant Proteins genetics, Recombinant Proteins metabolism, Recombinant Proteins pharmacology, Sulfones pharmacology, Gene Products, tax metabolism, Human T-lymphotropic virus 1 genetics, I-kappa B Kinase metabolism, Ubiquitination
- Abstract
Activation of NF-κB by human T cell leukaemia virus type 1 Tax is thought to be crucial in T-cell transformation and the onset of adult T cell leukaemia. Tax activates NF-κB through activation of the IκB kinase (IKK) complex, similar to cytokine-induced NF-κB activation, which involves active signalling complex formation using polyubiquitin chains as a platform. Although polyubiquitination of Tax was reported to be required for IKK activation, most studies have been performed using intact cells, in which secondary NF-κB activation can be induced by various cytokines that are secreted due to Tax-mediated primary NF-κB activation. Therefore, a cell-free assay system, in which IKK can be activated by adding highly purified recombinant Tax to cytosolic extract, was used to analyse Tax-induced IKK activation. In contrast to the cytosolic extract, the purified IKK complex was not activated by Tax, whereas, it was efficiently activated by MEKK1, that does not require polyubiquitination to activate IKK. Moreover, Tax-induced IKK activation was blocked when the cytosolic extract was mixed with either lysine-free, methylated or K63R ubiquitin. These results obtained through our cell-free assay suggest that K63-linked polyubiquitination is critical, but linear polyubiquitination is dispensable or insufficient for Tax-induced IKK activation.
- Published
- 2011
- Full Text
- View/download PDF
31. An artificial copper complex incorporating a cell-penetrating peptide inhibits nuclear factor-κB (NF-κB) activation.
- Author
-
Kanemaru Y, Momiki Y, Matsuura S, Horikawa T, Gohda J, Inoue J, Okamoto Y, Fujita M, and Otsuka M
- Subjects
- Coordination Complexes chemistry, Coordination Complexes pharmacology, HeLa Cells, Histamine chemistry, Histamine pharmacology, Humans, NF-kappa B immunology, tat Gene Products, Human Immunodeficiency Virus chemistry, tat Gene Products, Human Immunodeficiency Virus pharmacology, Cell-Penetrating Peptides chemistry, Cell-Penetrating Peptides pharmacology, Copper chemistry, Copper pharmacology, Histamine analogs & derivatives, NF-kappa B antagonists & inhibitors, Pyridines chemistry, Pyridines pharmacology
- Abstract
Nuclear factor-κB (NF-κB) is an inducible transcription factor activated by a variety of cytokines, and promotes the transcription of genes involved in cancer, inflammation, autoimmune disease, and viral infection, among others. Because of its involvement in numerous disease processes, considerable research has focused on NF-κB as a potential drug target. We previously reported that cupric ion (Cu(2+)) blocks NF-κB activation. However, Cu(2+) is unsuitable for drug applications. The copper complex of an artificial peptide HPH-Pep (HPH-Pep-Cu(2+)) was a promising alternative, but it did not easily cross the cell membrane. We report the development of a NF-κB inhibiting Cu(2+) complex with improved cell-penetrating activity arising from the coupling of a Tat peptide to HPH-Pep-Cu(2+).
- Published
- 2011
- Full Text
- View/download PDF
32. Human lactoferrin activates NF-kappaB through the Toll-like receptor 4 pathway while it interferes with the lipopolysaccharide-stimulated TLR4 signaling.
- Author
-
Ando K, Hasegawa K, Shindo K, Furusawa T, Fujino T, Kikugawa K, Nakano H, Takeuchi O, Akira S, Akiyama T, Gohda J, Inoue J, and Hayakawa M
- Subjects
- Animals, Cattle, Cell Line, Humans, Lipopolysaccharides immunology, Mice, Toll-Like Receptor 4 deficiency, Toll-Like Receptor 4 immunology, Lactoferrin metabolism, NF-kappa B metabolism, Signal Transduction, Toll-Like Receptor 4 metabolism
- Abstract
Lactoferrin (LF) has been implicated in innate immunity. Here we reveal the signal transduction pathway responsible for human LF (hLF)-triggered nuclear factor-kappaB (NF-kappaB) activation. Endotoxin-depleted hLF induces NF-kappaB activation at physiologically relevant concentrations in the human monocytic leukemia cell line, THP-1, and in mouse embryonic fibroblasts (MEFs). In MEFs, in which both tumor necrosis factor receptor-associated factor 2 (TRAF2) and TRAF5 are deficient, hLF causes NF-kappaB activation at a level comparable to that seen in wild-type MEFs, whereas TRAF6-deficient MEFs show significantly impaired NF-kappaB activation in response to hLF. TRAF6 is known to be indispensable in leading to NF-kappaB activation in myeloid differentiating factor 88 (MyD88)-dependent signaling pathways, while the role of TRAF6 in the MyD88-independent signaling pathway has not been clarified extensively. When we examined the hLF-dependent NF-kappaB activation in MyD88-deficient MEFs, delayed, but remarkable, NF-kappaB activation occurred as a result of the treatment of cells with hLF, indicating that both MyD88-dependent and MyD88-independent pathways are involved. Indeed, hLF fails to activate NF-kappaB in MEFs lacking Toll-like receptor 4 (TLR4), a unique TLR group member that triggers both MyD88-depependent and MyD88-independent signalings. Importantly, the carbohydrate chains from hLF are shown to be responsible for TLR4 activation. Furthermore, we show that lipopolysaccharide-induced cytokine and chemokine production is attenuated by intact hLF but not by the carbohydrate chains from hLF. Thus, we present a novel model concerning the biological function of hLF: hLF induces moderate activation of TLR4-mediated innate immunity through its carbohydrate chains; however, hLF suppresses endotoxemia by interfering with lipopolysaccharide-dependent TLR4 activation, probably through its polypeptide moiety.
- Published
- 2010
- Full Text
- View/download PDF
33. A unique domain in RANK is required for Gab2 and PLCgamma2 binding to establish osteoclastogenic signals.
- Author
-
Taguchi Y, Gohda J, Koga T, Takayanagi H, and Inoue J
- Subjects
- Amino Acid Sequence, Animals, CD40 Antigens metabolism, Conserved Sequence, Humans, Mice, Mice, Inbred C57BL, Models, Biological, Molecular Sequence Data, NFATC Transcription Factors metabolism, Protein Structure, Tertiary, Sequence Alignment, Transcriptional Activation, Adaptor Proteins, Signal Transducing metabolism, Cell Differentiation, Osteoclasts cytology, Phospholipase C gamma metabolism, Receptor Activator of Nuclear Factor-kappa B genetics, Receptor Activator of Nuclear Factor-kappa B metabolism, Signal Transduction
- Abstract
TRAF6 is essential for osteoclastogenesis and for both RANK- and CD40-mediated activation of IKK and MAPKs. RANK, but not CD40, can promote osteoclastogenesis because only RANK induces NFATc1 activation through PLCgamma2-induced Ca(2+) oscillations together with the co-stimulatory signals emanating from immune receptors linked to ITAM-containing adaptors. These previous data suggest that RANK harbors a unique domain that functions in concert with the TRAF6-binding site in osteoclastogenesis. Here we identify such a domain, highly conserved domain in RANK (HCR), which is dispensable for the early phase of RANK and ITAM signaling but is essential for their late-phase signaling, including sustained activation of NF-kappaB and PLCgamma2 leading to NFATc1 activation. HCR recruits an adaptor protein, Gab2, which further associates with PLCgamma2 in the late phase. Formation of the HCR-mediated signaling complex could account for the sustained activation of NF-kappaB and PLCgamma2. The present study identifies HCR as a unique domain that plays a critical role in the long-term linkage between RANK and ITAM signals, providing a molecular basis for therapeutic strategies.
- Published
- 2009
- Full Text
- View/download PDF
34. Two mechanistically and temporally distinct NF-kappaB activation pathways in IL-1 signaling.
- Author
-
Yamazaki K, Gohda J, Kanayama A, Miyamoto Y, Sakurai H, Yamamoto M, Akira S, Hayashi H, Su B, and Inoue J
- Subjects
- Animals, Cell Line, Humans, MAP Kinase Kinase Kinase 3 deficiency, MAP Kinase Kinase Kinase 3 metabolism, MAP Kinase Kinase Kinases deficiency, MAP Kinase Kinase Kinases genetics, MAP Kinase Kinase Kinases metabolism, Mice, Protein Binding, TNF Receptor-Associated Factor 6 deficiency, TNF Receptor-Associated Factor 6 metabolism, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitination, Interleukin-1 metabolism, NF-kappa B metabolism, Signal Transduction
- Abstract
The cytokine interleukin-1 (IL-1) mediates immune and inflammatory responses by activating the transcription factor nuclear factor kappaB (NF-kappaB). Although transforming growth factor-beta-activated kinase 1 (TAK1) and mitogen-activated protein kinase (MAPK) kinase kinase 3 (MEKK3) are both crucial for IL-1-dependent activation of NF-kappaB, their potential functional and physical interactions remain unclear. Here, we showed that TAK1-mediated activation of NF-kappaB required the transient formation of a signaling complex that included tumor necrosis factor receptor-associated factor 6 (TRAF6), MEKK3, and TAK1. Site-specific, lysine 63-linked polyubiquitination of TAK1 at lysine 209, likely catalyzed by TRAF6 and Ubc13, was required for the formation of this complex. After TAK1-mediated activation of NF-kappaB, TRAF6 subsequently activated NF-kappaB through MEKK3 independently of TAK1, thereby establishing continuous activation of NF-kappaB, which was required for the production of sufficient cytokines. Therefore, we propose that the cooperative activation of NF-kappaB by two mechanistically and temporally distinct MEKK3-dependent pathways that diverge at TRAF6 critically contributes to immune and inflammatory systems.
- Published
- 2009
- Full Text
- View/download PDF
35. NIK is involved in constitutive activation of the alternative NF-kappaB pathway and proliferation of pancreatic cancer cells.
- Author
-
Nishina T, Yamaguchi N, Gohda J, Semba K, and Inoue J
- Subjects
- Cell Line, Tumor, Gene Silencing, Humans, Pancreatic Neoplasms enzymology, Protein Serine-Threonine Kinases genetics, NF-kappaB-Inducing Kinase, Cell Proliferation, NF-kappa B p52 Subunit metabolism, Pancreatic Neoplasms pathology, Protein Serine-Threonine Kinases metabolism, Transcription Factor RelB metabolism
- Abstract
Pancreatic cancer has one of the poorest prognoses among human neoplasms. Constitutive activation of NF-kappaB is frequently observed in pancreatic cancer cells and is involved in their malignancy. However, little is known about the molecular mechanism of this constitutive NF-kappaB activation. Here, we show that the alternative pathway is constitutively activated and NF-kappaB-inducing kinase (NIK), a mediator of the alternative pathway, is significantly expressed in pancreatic cancer cells. siRNA-mediated silencing of NIK expression followed by subcellular fractionation revealed that NIK is constitutively involved in the processing of p100 and nuclear transport of p52 and RelB in pancreatic cancer cells. In addition, NIK silencing significantly suppressed proliferation of pancreatic cancer cells. These results clearly indicate that NIK is involved in the constitutive activation of the alternative pathway and controls cell proliferation in pancreatic cancer cells. Therefore, NIK might be a novel target for the treatment of pancreatic cancer.
- Published
- 2009
- Full Text
- View/download PDF
36. TRAF6 establishes innate immune responses by activating NF-kappaB and IRF7 upon sensing cytosolic viral RNA and DNA.
- Author
-
Konno H, Yamamoto T, Yamazaki K, Gohda J, Akiyama T, Semba K, Goto H, Kato A, Yujiri T, Imai T, Kawaguchi Y, Su B, Takeuchi O, Akira S, Tsunetsugu-Yokota Y, and Inoue J
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Cell Line, Cytosol enzymology, Cytosol immunology, DEAD-box RNA Helicases metabolism, Humans, Interferon Regulatory Factor-3 metabolism, MAP Kinase Kinase Kinase 3 metabolism, MAP Kinase Kinase Kinases metabolism, Mice, Protein Binding, Protein Serine-Threonine Kinases metabolism, RNA Viruses isolation & purification, Signal Transduction, Cytosol virology, DNA, Viral immunology, Immunity, Innate immunology, Interferon Regulatory Factor-7 metabolism, NF-kappa B metabolism, RNA, Viral immunology, TNF Receptor-Associated Factor 6 metabolism
- Abstract
Background: In response to viral infection, the innate immune system recognizes viral nucleic acids and then induces production of proinflammatory cytokines and type I interferons (IFNs). Toll-like receptor 7 (TLR7) and TLR9 detect viral RNA and DNA, respectively, in endosomal compartments, leading to the activation of nuclear factor kappaB (NF-kappaB) and IFN regulatory factors (IRFs) in plasmacytoid dendritic cells. During such TLR signaling, TNF receptor-associated factor 6 (TRAF6) is essential for the activation of NF-kappaB and the production of type I IFN. In contrast, RIG-like helicases (RLHs), cytosolic RNA sensors, are indispensable for antiviral responses in conventional dendritic cells, macrophages, and fibroblasts. However, the contribution of TRAF6 to the detection of cytosolic viral nucleic acids has been controversial, and the involvement of TRAF6 in IRF activation has not been adequately addressed., Principal Findings: Here we first show that TRAF6 plays a critical role in RLH signaling. The absence of TRAF6 resulted in enhanced viral replication and a significant reduction in the production of IL-6 and type I IFNs after infection with RNA virus. Activation of NF-kappaB and IRF7, but not that of IRF3, was significantly impaired during RLH signaling in the absence of TRAF6. TGFbeta-activated kinase 1 (TAK1) and MEKK3, whose activation by TRAF6 during TLR signaling is involved in NF-kappaB activation, were not essential for RLH-mediated NF-kappaB activation. We also demonstrate that TRAF6-deficiency impaired cytosolic DNA-induced antiviral responses, and this impairment was due to defective activation of NF-kappaB and IRF7., Conclusions/significance: Thus, TRAF6 mediates antiviral responses triggered by cytosolic viral DNA and RNA in a way that differs from that associated with TLR signaling. Given its essential role in signaling by various receptors involved in the acquired immune system, TRAF6 represents a key molecule in innate and antigen-specific immune responses against viral infection.
- Published
- 2009
- Full Text
- View/download PDF
37. Predicting novel features of toll-like receptor 3 signaling in macrophages.
- Author
-
Helmy M, Gohda J, Inoue J, Tomita M, Tsuchiya M, and Selvarajoo K
- Subjects
- Animals, Mice, Macrophages metabolism, Signal Transduction, Toll-Like Receptor 3 metabolism
- Abstract
The Toll-like receptor (TLR) 3 plays a critical role in mammalian innate immune response against viral attacks by recognizing double-stranded RNA (dsRNA) or its synthetic analog polyinosinic-polycytidylic acid (poly (IratioC)). This leads to the activation of MAP kinases and NF-kappaB which results in the induction of type I interferons and proinflammatory cytokines to combat the viral infection. To understand the complex interplay of the various intracellular signaling molecules in the regulation of NF-kappaB and MAP kinases, we developed a computational TLR3 model based upon perturbation-response approach. We curated literature and databases to determine the TLR3 signaling topology specifically for murine macrophages. For initial model creation, we used wildtype temporal activation profiles of MAP kinases and NF-kappaB and, for model testing, used TRAF6 KO and TRADD KO data. From dynamic simulations we predict i) the existence of missing intermediary steps between extracellular poly (IratioC) stimulation and intracellular TLR3 binding, and ii) the presence of a novel pathway which is essential for JNK and p38, but not NF-kappaB, activation. Our work shows activation dynamics of signaling molecules can be used in conjunction with perturbation-response models to decipher novel signaling features of complicated immune pathways.
- Published
- 2009
- Full Text
- View/download PDF
38. Signaling flux redistribution at toll-like receptor pathway junctions.
- Author
-
Selvarajoo K, Takada Y, Gohda J, Helmy M, Akira S, Tomita M, Tsuchiya M, Inoue J, and Matsuo K
- Subjects
- Animals, Cells, Cultured, Computational Biology, Humans, Interferon Regulatory Factor-3 metabolism, Mice, Mice, Knockout, Myeloid Differentiation Factor 88 genetics, Myeloid Differentiation Factor 88 metabolism, Receptors, Interleukin genetics, Receptors, Interleukin metabolism, TNF Receptor-Associated Factor 6 genetics, TNF Receptor-Associated Factor 6 metabolism, Transfection, Signal Transduction, Toll-Like Receptor 4 metabolism
- Abstract
Various receptors on cell surface recognize specific extracellular molecules and trigger signal transduction altering gene expression in the nucleus. Gain or loss-of-function mutations of one molecule have shown to affect alternative signaling pathways with a poorly understood mechanism. In Toll-like receptor (TLR) 4 signaling, which branches into MyD88- and TRAM-dependent pathways upon lipopolysaccharide (LPS) stimulation, we investigated the gain or loss-of-function mutations of MyD88. We predict, using a computational model built on the perturbation-response approach and the law of mass conservation, that removal and addition of MyD88 in TLR4 activation, enhances and impairs, respectively, the alternative TRAM-dependent pathway through signaling flux redistribution (SFR) at pathway branches. To verify SFR, we treated MyD88-deficient macrophages with LPS and observed enhancement of TRAM-dependent pathway based on increased IRF3 phosphorylation and induction of Cxcl10 and Ifit2. Furthermore, increasing the amount of MyD88 in cultured cells showed decreased TRAM binding to TLR4. Investigating another TLR4 pathway junction, from TRIF to TRAF6, RIP1 and TBK1, the removal of MyD88-dependent TRAF6 increased expression of TRAM-dependent Cxcl10 and Ifit2. Thus, we demonstrate that SFR is a novel mechanism for enhanced activation of alternative pathways when molecules at pathway junctions are removed. Our data suggest that SFR may enlighten hitherto unexplainable intracellular signaling alterations in genetic diseases where gain or loss-of-function mutations are observed.
- Published
- 2008
- Full Text
- View/download PDF
39. HTLV-1 Tax-induced NFkappaB activation is independent of Lys-63-linked-type polyubiquitination.
- Author
-
Gohda J, Irisawa M, Tanaka Y, Sato S, Ohtani K, Fujisawa J, and Inoue J
- Subjects
- Humans, Jurkat Cells, Lysine chemistry, Ubiquitin chemistry, Human T-lymphotropic virus 1 metabolism, I-kappa B Kinase metabolism, Lysine metabolism, NF-kappa B metabolism, Signal Transduction physiology, Ubiquitin metabolism
- Abstract
Human T-cell leukemia virus type 1 (HTLV-1) Tax-induced activation of nuclear factor-kappaB (NFkappaB) is thought to play a critical role in T-cell transformation and onset of adult T-cell leukemia. However, the molecular mechanism of the Tax-induced NFkappaB activation remains unknown. One of the mitogen-activated protein kinase kinase kinses (MAP3Ks) members, TAK1, plays a critical role in cytokine-induced activation of NFkappaB, which involves lysine 63-linked (K63) polyubiquitination of NEMO, a noncatalytic subunit of the IkappaB kinase complex. Here we show that Tax induces K63 polyubiquitination of NEMO. However, TAK1 is dispensable for Tax-induced NFkappaB activation, and deubiquitination of the K63 polyubiquitin chain failed to block Tax-induced NFkappaB activation. In addition, silencing of other MAP3Ks, including MEKK1, MEKK3, NIK, and TPL-2, did not affect Tax-induced NFkappaB activation. These results strongly suggest that unlike cytokine signaling, Tax-induced NFkappaB activation does not involve K63 polyubiquitination-mediated MAP3K activation.
- Published
- 2007
- Full Text
- View/download PDF
40. NF-kappaB activation in development and progression of cancer.
- Author
-
Inoue J, Gohda J, Akiyama T, and Semba K
- Subjects
- Binding Sites, Disease Progression, Humans, Models, Biological, Neoplasms pathology, Osteoclasts metabolism, RANK Ligand metabolism, TNF Receptor-Associated Factor 6 metabolism, NF-kappa B metabolism, Neoplasms metabolism
- Abstract
Nuclear factor-kappaBeta (NF-kappaB) binds specifically to NF-kappaB-binding sites (kappaB sites, 5'-GGGRNNYYCC-3'; R, purine; Y, pyrimidine; N, any nucleotide) present in enhancer regions of various genes. Binding of various cytokines, growth factors and pathogen-associated molecular patterns to specific receptors activates NF-kappaB and expression of genes that play critical roles in inflammation, innate and acquired immunity, bone remodeling and generation of skin appendices. Activation of NF-kappaB is also involved in cancer development and progression. NF-kappaB is activated in cells that become malignant tumors and in cells that are recruited to and constitute the tumor microenvironment. In the latter scenario, the TLR-TRAF6-NF-kB pathways seem to play major roles, and NF-kappaB activation results in production of cytokines, which in turn induce NF-kappaB activation in premalignant cells, leading to expression of genes involved abnormal growth and malignancy. Furthermore, NF-kappaB activation is involved in bone metastasis. Osteoclasts, whose generation requires the RANK-TRAF6-NF-kappaB pathways, release various growth factors stored in bone, which results in creation of microenvironment suitable for proliferation and colonization of cancer cells. Therefore, NF-kappaB and molecules involved its activation, such as TRAF6, are attractive targets for therapeutic strategies against cancer.
- Published
- 2007
- Full Text
- View/download PDF
41. Characteristics and biological functions of TRAF6.
- Author
-
Inoue J, Gohda J, and Akiyama T
- Subjects
- Animals, Humans, Signal Transduction physiology, TNF Receptor-Associated Factor 6 chemistry, TNF Receptor-Associated Factor 6 physiology
- Abstract
TRAF6 is divergent from other members of the TRAF family. Therefore, TRAF6 was expected to play physiological roles distinct from those of other TRAFs. In this chapter, we focused on the physiological functions specific to TRAF6 but not other TRAFs in immune system, formation of skin appendices, and nervous system development by describing abnormal phenotypes observed in TRAF6-deficient mice. The role of TRAF6 in osteoclastogenesis and the molecular mechanisms ofTRAF6-mediated signal transduction are described in other chapters.
- Published
- 2007
- Full Text
- View/download PDF
42. Recruitment of tumor necrosis factor receptor-associated factor family proteins to apoptosis signal-regulating kinase 1 signalosome is essential for oxidative stress-induced cell death.
- Author
-
Noguchi T, Takeda K, Matsuzawa A, Saegusa K, Nakano H, Gohda J, Inoue J, and Ichijo H
- Subjects
- Animals, Cells, Cultured, Fibroblasts cytology, Fibroblasts metabolism, Humans, Immunoprecipitation, Intracellular Signaling Peptides and Proteins metabolism, Kidney metabolism, Macrophages cytology, Macrophages metabolism, Membrane Proteins, Mice, Mice, Knockout, Spleen cytology, Spleen metabolism, Apoptosis, Oxidative Stress, Signal Transduction, TNF Receptor-Associated Factor 2 metabolism, TNF Receptor-Associated Factor 6 metabolism
- Abstract
Apoptosis signal-regulating kinase 1 (ASK1) plays a pivotal role in oxidative stress-induced cell death. Reactive oxygen species disrupt the interaction of ASK1 with its cellular inhibitor thioredoxin and thereby activates ASK1. However, the precise mechanism by which ASK1 freed from thioredoxin undergoes oligomerization-dependent activation has not been fully elucidated. Here we show that endogenous ASK1 constitutively forms a high molecular mass complex including Trx ( approximately 1,500-2,000 kDa), which we designate ASK1 signalosome. Upon H(2)O(2) treatment, the ASK1 signalosome forms a higher molecular mass complex at least in part because of the recruitment of tumor necrosis factor receptor-associated factor 2 (TRAF2) and TRAF6. Consistent with our previous findings that TRAF2 and TRAF6 activate ASK1, H(2)O(2)-induced ASK1 activation and cell death were strongly reduced in the cells derived from Traf2-/- and Traf6-/- mice. A novel signaling complex including TRAF2, TRAF6, and ASK1 may thus be the key component in oxidative stress-induced cell death.
- Published
- 2005
- Full Text
- View/download PDF
43. RANK-mediated amplification of TRAF6 signaling leads to NFATc1 induction during osteoclastogenesis.
- Author
-
Gohda J, Akiyama T, Koga T, Takayanagi H, Tanaka S, and Inoue J
- Subjects
- Animals, Binding Sites, CD40 Antigens chemistry, CD40 Antigens metabolism, Carrier Proteins genetics, Cells, Cultured, Cytoplasm metabolism, Humans, Membrane Glycoproteins genetics, Mice, NF-kappa B metabolism, NFATC Transcription Factors, Protein Binding, RANK Ligand, Receptor Activator of Nuclear Factor-kappa B, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Carrier Proteins metabolism, DNA-Binding Proteins metabolism, Membrane Glycoproteins metabolism, Nuclear Proteins metabolism, Osteoclasts cytology, Osteoclasts metabolism, Signal Transduction, TNF Receptor-Associated Factor 6 metabolism, Transcription Factors metabolism
- Abstract
RANK and CD40 activate NF-kappaB and MAPKs to similar levels via TRAF6. Even though overexpression of TRAF6 results in osteoclast formation, RANK but not CD40 promotes osteoclastogenesis. To understand the molecular basis for RANK-specific activity in osteoclastogenesis, we created an osteoclast formation system driven by anti-human CD40 antibody-mediated stimulation of a chimeric receptor, h40/mRK, which consists of the extracellular domain of human CD40 and the transmembrane and cytoplasmic domains of mouse RANK. By introducing mutations into three TRAF6-binding sites of RANK, we found that h40/mRK with a single TRAF6-binding site efficiently induced Ca2+ oscillation and expression of NFATc1, a master switch in osteoclastogenesis, whereas CD40 carrying a single TRAF6-binding site did not. However, expression of CD40 that was approximately 100 times greater than that of h40/mRK resulted in osteoclast formation, indicating that the RANK-TRAF6 signal is more potent than the CD40-TRAF6 signal in terms of NFATc1 activation and osteoclastogenesis. These results suggest that RANK may harbor a specific domain that amplifies TRAF6 signaling.
- Published
- 2005
- Full Text
- View/download PDF
44. Cutting edge: TNFR-associated factor (TRAF) 6 is essential for MyD88-dependent pathway but not toll/IL-1 receptor domain-containing adaptor-inducing IFN-beta (TRIF)-dependent pathway in TLR signaling.
- Author
-
Gohda J, Matsumura T, and Inoue J
- Subjects
- Adaptor Proteins, Signal Transducing, Adaptor Proteins, Vesicular Transport genetics, Animals, Humans, Macrophages metabolism, Membrane Glycoproteins genetics, Mice, Myeloid Differentiation Factor 88, Proteins genetics, Receptors, Interleukin-1 genetics, Receptors, Interleukin-1 metabolism, TNF Receptor-Associated Factor 6, Toll-Like Receptor 2, Toll-Like Receptor 3, Toll-Like Receptor 4, Toll-Like Receptor 5, Toll-Like Receptor 7, Toll-Like Receptor 9, Toll-Like Receptors, Adaptor Proteins, Vesicular Transport metabolism, Antigens, Differentiation metabolism, Membrane Glycoproteins metabolism, Proteins metabolism, Receptors, Cell Surface metabolism, Receptors, Immunologic metabolism, Signal Transduction physiology
- Abstract
Signaling pathways from TLRs are mediated by the Toll/IL-1R (TIR) domain-containing adaptor molecules. TNF receptor-associated factor (TRAF) 6 is thought to activate NF-kappaB and MAPKs downstream of these TIR domain-containing proteins to induce production of inflammatory cytokines. However, the precise role of TRAF6 in signaling from individual TLRs has not been appropriately addressed. We analyzed macrophages from TRAF6-deficient mice and made the following observations. In the absence of TRAF6, 1) ligands for TLR2, TLR5, TLR7, and TLR9 failed to induce activation of NF-kappaB and MAPKs or production of inflammatory cytokines; 2) TLR4 ligand-induced cytokine production was remarkably reduced and activation of NF-kappaB and MAPKs was observed, albeit with delayed kinetics; and 3) in contrast with previously reported findings, TLR3 signaling was not affected. These results indicate that TRAF6 is essential for MyD88-dependent signaling but is not required for TIR domain-containing adaptor-inducing IFN-beta (TRIF)-dependent signaling.
- Published
- 2004
- Full Text
- View/download PDF
45. TIFAB inhibits TIFA, TRAF-interacting protein with a forkhead-associated domain.
- Author
-
Matsumura T, Semba K, Azuma S, Ikawa S, Gohda J, Akiyama T, and Inoue J
- Subjects
- Amino Acid Sequence, Animals, Blotting, Western, Cell Line, Gene Expression, Humans, Mice, Molecular Sequence Data, NF-kappa B antagonists & inhibitors, NF-kappa B metabolism, Precipitin Tests, RNA, Messenger biosynthesis, Receptors, Tumor Necrosis Factor antagonists & inhibitors, Receptors, Tumor Necrosis Factor chemistry, Receptors, Tumor Necrosis Factor genetics, TNF Receptor-Associated Factor 6, Transfection, Tumor Necrosis Factor Receptor-Associated Peptides and Proteins, Proteins antagonists & inhibitors, Receptors, Tumor Necrosis Factor metabolism
- Abstract
Tumor necrosis factor receptor-associated factor 6 (TRAF6) transduces signals that lead to activation of NFkappaB and AP-1, which is essential for cell differentiation and establishment of the immune and inflammatory systems. TRAF-interacting protein with a forkhead-associated domain (TIFA) was identified as a TRAF6-binding protein that could link IRAK-1 to TRAF6 and then activate TRAF6 upon stimulation. We report identification of a TIFA-related protein, TIFAB, that inhibits TIFA-mediated activation of NFkappaB. TIFAB does not associate with members of the TRAF family but does bind TIFA. We analyzed the effect of TIFAB expression on the TRAF6/TIFA interaction by immunoprecipitation of TRAF6 and found that TIFA coprecipitated with TRAF6 was not changed. However, when we analyzed this interaction by immunoprecipitation of TIFA, we found that TIFAB significantly increased the amount of TRAF6 coprecipitated with TIFA. These findings suggest that TIFAB inhibits the TIFA-mediated TRAF6 activation possibly by inducing a conformational change in TIFA.
- Published
- 2004
- Full Text
- View/download PDF
46. Cloning and characterization of Xenopus laevis drg2, a member of the developmentally regulated GTP-binding protein subfamily.
- Author
-
Ishikawa K, Azuma S, Ikawa S, Morishita Y, Gohda J, Akiyama T, Semba K, and Inoue Ji
- Subjects
- Amino Acid Sequence, Animals, Blotting, Northern, Cloning, Molecular, DNA, Complementary chemistry, DNA, Complementary genetics, Embryo, Nonmammalian metabolism, Embryonic Development, Female, GTP-Binding Proteins metabolism, Gene Expression Regulation, Developmental, In Situ Hybridization, Molecular Sequence Data, Protein Binding, RNA metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Sequence Alignment, Sequence Analysis, DNA, Sequence Homology, Amino Acid, Xenopus Proteins metabolism, Xenopus laevis embryology, Xenopus laevis growth & development, GTP-Binding Proteins genetics, Xenopus Proteins genetics, Xenopus laevis genetics
- Abstract
The developmentally regulated GTP-binding protein (DRG) subfamily is an uncharacterized member of the Obg family, an evolutional branch of GTPase superfamily proteins. GTPases act as molecular switches regulating diverse cellular processes. DRG2 and DRG1 comprise the DRG subfamily in eucaryotes. Although drg1 was first identified as a gene predominantly expressed during early development of the mouse central nervous system, comparative analysis of drg2 and drg1 expression during embryogenesis has never been reported, and the biochemical properties of the DRG family proteins remain to be elucidated. Thus, we first cloned Xenopus drg2 (Xdrg2) and examined the temporal and spatial expression patterns of Xdrg2 mRNA in comparison to those of Xdrg1. Both Xdrg2 and Xdrg1 are induced at late gastrula and subsequently increased during later stages of embryos (stage 13-41). Whole-mount in situ hybridization showed that Xdrg2 and Xdrg1 expression patterns are almost identical except that only Xdrg2 expression is detected in the stage 22 pronephric anlage. Strong transcripts of both genes are also observed at this stage in neural crest cells, blood islands, and developing eyes, and in brain, eyes, otic vesicle, branchial arches, pronephroses, spinal cord, notochord, head mesenchyme, and somites at stages 27 and 32. Northern blot analysis of adult tissues revealed that both genes are expressed highly in ovary and testis and rather moderately in other organs, except that Xdrg1 transcripts are scarcely detected in heart, lung, and liver. Accordingly, transcription or stability of Xdrg2 and Xdrg1 mRNAs may be regulated by different mechanisms. In addition, by generating recombinant XDRG2 and XDRG1 proteins, we found the RNA binding activity of these proteins in vitro. Our results suggest that the DRG proteins may play their physiological roles via RNA binding.
- Published
- 2003
- Full Text
- View/download PDF
47. A tetraspanin-family protein, T-cell acute lymphoblastic leukemia-associated antigen 1, is induced by the Ewing's sarcoma-Wilms' tumor 1 fusion protein of desmoplastic small round-cell tumor.
- Author
-
Ito E, Honma R, Imai J, Azuma S, Kanno T, Mori S, Yoshie O, Nishio J, Iwasaki H, Yoshida K, Gohda J, Inoue J, Watanabe S, and Semba K
- Subjects
- Cell Line, Tumor, Gene Expression Profiling, Humans, Membrane Glycoproteins genetics, Oligonucleotide Array Sequence Analysis, Oncogene Proteins, Fusion genetics, RNA, Messenger metabolism, Transfection, Carcinoma, Small Cell metabolism, Membrane Glycoproteins metabolism, Oncogene Proteins, Fusion metabolism, Peritoneal Neoplasms metabolism
- Abstract
Recurrent chromosomal translocations in neoplasms often generate hybrid genes that play critical roles in tumorigenesis. Desmoplastic small round-cell tumor (DSRCT) is an aggressive malignancy associated with the chromosomal translocation t(11;22)(p13;q12). This translocation generates a chimeric transcription factor, EWS-WT1, which consists of the transcriptional activation domain of the Ewing's sarcoma (EWS) protein and the DNA binding domain of the Wilms' tumor 1 (WT1) protein. One of the splice variants, EWS-WT1(-KTS) lacks three amino acid residues (Lys-Thr-Ser) in the DNA binding domain and transforms NIH3T3 cells. Therefore, it is likely that aberrant gene expression caused by EWS-WT1(-KTS) is involved in the malignant phenotype of DSRCT. Microarray analysis of 9600 human genes revealed that a gene encoding a tetraspanin-family protein, T-cell acute lymphoblastic leukemia-associated antigen 1 (TALLA-1), was induced in EWS-WT1(-KTS)-expressing cell clones. This induction was EWS-WT1(-KTS)-specific, and more importantly, TALLA-1 protein was expressed in the three independent cases of DSRCT. Tetraspanin-family genes encode transmembrane proteins that regulate various cell processes such as cell adhesion, migration and metastasis. Our findings provide a novel insight into the malignant phenotype of DSRCT, suggesting that TALLA-1 is a useful marker for diagnosis and a potential target for the therapy of DSRCT.
- Published
- 2003
- Full Text
- View/download PDF
48. Down-regulation of TNF-alpha receptors by conophylline in human T-cell leukemia cells.
- Author
-
Gohda J, Inoue J, and Umezawa K
- Subjects
- Antigens, CD biosynthesis, Binding Sites, Cell Membrane metabolism, Cell Nucleus metabolism, Cell Separation, Dose-Response Relationship, Drug, Enzyme Activation, Flow Cytometry, HL-60 Cells, Humans, I-kappa B Proteins metabolism, Jurkat Cells, Kinetics, Models, Chemical, NF-KappaB Inhibitor alpha, NF-kappa B metabolism, Phosphorylation, Protein Binding, Receptors, Tumor Necrosis Factor, Type I, Recombinant Proteins metabolism, Time Factors, Down-Regulation, Leukemia, T-Cell drug therapy, Leukemia, T-Cell metabolism, Receptors, Tumor Necrosis Factor biosynthesis, Vinca Alkaloids pharmacology
- Abstract
In the course of our screening for tumor necrosis factor-alpha (TNF-alpha) function inhibitors, conophylline, a vinca alkaloid isolated from the plant Ervatamia microphylla, was found to inhibit TNF-alpha-induced NF-kappaB activation. We studied the effect of conophylline on TNF-alpha-induced NF-kappaB and JNK activations in human T-cell leukemia Jurkat cells. Conophylline inhibited both of these TNF-alpha-induced activations. It also inhibited phosphorylation and degradation of I-kappaB-alpha. Moreover, a receptor binding assay using [125I]-TNF-alpha showed that this inhibitory effect was due to a decrease in the binding of TNF-alpha to the cells. Scatchard analysis of the binding data indicated that conophylline induced only a small change in the affinity of the receptors but a significant change in the receptor number. FACS analysis showed that conophylline reduced the expression of CD120a/TNFR1, the high-affinity receptor for TNF-alpha, on the cell surface. On the other hand, conophylline did not affect the kinetics of internalization and degradation of TNF-alpha/receptor complexes or the half-life of TNF-alpha binding sites. These results indicate that conophylline down-regulates the expression of the TNF-alpha receptors on the cell surface.
- Published
- 2003
49. Identification of TIFA as an adapter protein that links tumor necrosis factor receptor-associated factor 6 (TRAF6) to interleukin-1 (IL-1) receptor-associated kinase-1 (IRAK-1) in IL-1 receptor signaling.
- Author
-
Takatsuna H, Kato H, Gohda J, Akiyama T, Moriya A, Okamoto Y, Yamagata Y, Otsuka M, Umezawa K, Semba K, and Inoue J
- Subjects
- Animals, Female, Interleukin-1 pharmacology, Interleukin-1 Receptor-Associated Kinases, Mice, Mice, Inbred ICR, Molecular Sequence Data, NF-kappa B metabolism, Receptors, Interleukin-1 metabolism, Sequence Homology, Amino Acid, Signal Transduction drug effects, Signal Transduction physiology, TNF Receptor-Associated Factor 6, Adaptor Proteins, Signal Transducing, Carrier Proteins genetics, Carrier Proteins metabolism, Protein Kinases metabolism, Proteins metabolism, Receptors, Tumor Necrosis Factor metabolism
- Abstract
Tumor necrosis factor receptor-associated factor 6 (TRAF6) transduces signals from members of the Toll/interleukin-1 (IL-1) receptor family by interacting with IL-1 receptor-associated kinase-1 (IRAK-1) after IRAK-1 is released from the receptor-MyD88 complex upon IL-1 stimulation. However, the molecular mechanisms underlying regulation of the IRAK-1/TRAF6 interaction are largely unknown. We have identified TIFA, a TRAF-interacting protein with a forkhead-associated (FHA) domain. The FHA domain is a motif known to bind directly to phosphothreonine and phosphoserine. In transient transfection assays, TIFA activates NFkappaBeta and c-Jun amino-terminal kinase. However, TIFA carrying a mutation that abolishes TRAF6 binding or mutations in the FHA domain that are known to abolish FHA domain binding to phosphopeptide fails to activate NFkappaBeta and c-Jun amino-terminal kinase. TIFA, when overexpressed, binds both TRAF6 and IRAK-1 and significantly enhances the IRAK-1/TRAF6 interaction. Furthermore, analysis of endogenous proteins indicates that TIFA associates with TRAF6 constitutively, whereas it associates with IRAK-1 in an IL-1 stimulation-dependent manner in vivo. Thus, TIFA is likely to mediate IRAK-1/TRAF6 interaction upon IL-1 stimulation.
- Published
- 2003
- Full Text
- View/download PDF
50. Elimination of the vertebrate Escherichia coli Ras-like protein homologue leads to cell cycle arrest at G1 phase and apoptosis.
- Author
-
Gohda J, Nomura Y, Suzuki H, Arai H, Akiyama T, and Inoue J
- Subjects
- Animals, Cell Division, Cell Line cytology, Chickens metabolism, Consensus Sequence, Doxycycline pharmacology, GTP-Binding Proteins deficiency, GTP-Binding Proteins genetics, Gene Targeting, Genes, Reporter, Luciferases biosynthesis, Luciferases genetics, Prokaryotic Cells metabolism, Promoter Regions, Genetic drug effects, Protein Structure, Tertiary, Proto-Oncogene Proteins c-bcl-2 genetics, RNA-Binding Proteins genetics, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins genetics, Transfection, Vertebrates metabolism, bcl-X Protein, Apoptosis physiology, G1 Phase physiology, GTP-Binding Proteins physiology, RNA-Binding Proteins physiology
- Abstract
Homologues of the Escherichia coli (E. coli) Ras-like protein (ERA), a GTP-binding protein with RNA binding activity, have recently been found in various species, including human, mouse, and Antirrhinum majus. Depletion of prokaryotic ERA blocks cell division without affecting chromosome segregation. However, the physiological function of eukaryotic ERA is largely unknown. We have performed a genetic analysis of chicken ERA (GdERA) in DT40 cells. Depletion of GdERA diminished the growth rate of the cells, accompanied by an accumulation of apoptotic cells. The analysis of cell cycle indicates that the elimination of GdERA caused arrest at G1 phase, but not at M phase, which highlights the distinct role of vertebrate ERA in the cell cycle progression compared to prokaryotic ERA. Furthermore, human ERA (HsERA) rescued the phenotype of GdERA-deficient cells, whereas a mutant of HsERA deprived of RNA-binding activity did not. These data suggest that vertebrate ERA regulates the G1 phase progression via an as yet unknown molecular mechanism, which involves RNA recognition by ERA.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.