1
|
Jacob JR, Singh R, Okamoto M, Chakravarti A, Palanichamy K. miRNA-194-3p represses NF-κB in gliomas to attenuate iPSC genes and proneural to mesenchymal transition. iScience 2024; 27:108650. [PMID: 38226170 PMCID: PMC10788216 DOI: 10.1016/j.isci.2023.108650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/01/2023] [Accepted: 12/04/2023] [Indexed: 01/17/2024] Open
Abstract
Severe tumor heterogeneity drives the aggressive and treatment refractory nature of glioblastomas (GBMs). While limiting GBM heterogeneity offers promising therapeutic potential, the underlying mechanisms that regulate GBM plasticity remain poorly understood. We utilized 14 patient-derived and four commercially available cell lines to uncover miR-194-3p as a key epigenetic determinant of stemness and transcriptional subtype in GBM. We demonstrate that miR-194-3p degrades TAB2, an important mediator of NF-κB activity, decreasing NF-κB transcriptional activity. The loss in NF-κB activity following miR-194-3p overexpression or TAB2 silencing decreased expression of induced pluripotent stem cell (iPSC) genes, inhibited the oncogenic IL-6/STAT3 signaling axis, suppressed the mesenchymal transcriptional subtype in relation to the proneural subtype, and induced differentiation from the glioma stem cell (GSC) to monolayer (ML) phenotype. miR-194-3p/TAB2/NF-κB signaling axis acts as an epigenetic switch that regulates GBM plasticity and targeting this signaling axis represents a potential strategy to limit transcriptional heterogeneity in GBMs.
Collapse
Affiliation(s)
- John Ryan Jacob
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Rajbir Singh
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Masa Okamoto
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Kamalakannan Palanichamy
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
2
|
Luo J, Wu F, Liu W, Ren Q, Diao P, Guan G, Luo J, Yin H, Liu G. A Novel MicroRNA and the Target Gene TAB2 Can Regulate the Process of Sucking Blood in and the Spawn Rate of Hyalomma asiaticum (Acari: Ixodidae) Ticks. Front Immunol 2022; 13:930532. [PMID: 35865515 PMCID: PMC9294593 DOI: 10.3389/fimmu.2022.930532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/14/2022] [Indexed: 01/10/2023] Open
Abstract
Ticks are blood-sucking parasites that are harmful to humans and animals. MicroRNAs are a class of conserved small noncoding RNAs that play regulatory roles in the expression of many genes at the posttranscriptional level. Here, a novel miRNA (nov-miR-17) was identified from a small RNA data library of Hyalomma asiaticum by next-generation sequencing. PCR was used to obtain precursor nov-miR-17 by RACE using mature loop primers. The secondary structure was predicted with UNAFold. The interaction of nov-miR-17 with its target gene TAB2 was predicted using RNAhybrid software and identified in vitro by luciferase assays. Moreover, the interaction was confirmed in vivo by phenotype rescue experiments in which dsTAB2 was used for RNA interference (RNAi) and an antagomir of nov-miR-17 was used for miRNA silencing. The expression levels of nov-miR-17 and TAB2 in ticks at different developmental stages and the expression of nov-miR-17 in different tissues were analyzed by real-time qPCR. All data were analyzed using GraphPad Prism version 5. Results: The results showed that TAB2 was a target gene of nov-miR-17. When the blood-sucking process of larval, nymph and adult ticks was prolonged, the expression of nov-miR-17 was decreased, and TAB2 expression was increased. However, the level of nov-miR-17 in the midgut of engorged ticks was highest at all stages. Therefore, nov-miR-17 plays an important role in the blood-sucking process. The overexpression of nov-miR-17 indicated that this miRNA affected the engorged weight (P < 0.001) and spawn rate (P < 0.001) of female ticks. RNAi of TAB2 also had the same effect. dsRNA not only impacted the weight (P < 0.01) but also reduced the spawn rate (P < 0.001) of the ticks. Furthermore, significant recovery was observed in nov-miR-17-silenced ticks after TAB2 silencing by RNAi. nov-miR-17 silencing by antagomir not only impacted the engorged weight of the female ticks (P < 0.001) but also the number of days that the females needed to progress from engorgement to spawning (P < 0.001). The study showed that nov-miR-17, as a new miRNA, plays an important role along with its target gene TAB2 in the blood-sucking and spawning processes in female ticks.
Collapse
Affiliation(s)
- Jin Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, China
| | - Feng Wu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, China
| | - Wenge Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, China
| | - Qiaoyun Ren
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, China
| | - Peiwen Diao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, China
| | - Guiquan Guan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, China
| | - Jianxun Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, China
| | - Hong Yin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, China
- *Correspondence: Guangyuan Liu, ; Hong Yin,
| | - Guangyuan Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, China
- *Correspondence: Guangyuan Liu, ; Hong Yin,
| |
Collapse
|
3
|
Lerner L, Tao J, Liu Q, Nicoletti R, Feng B, Krieger B, Mazsa E, Siddiquee Z, Wang R, Huang L, Shen L, Lin J, Vigano A, Chiu MI, Weng Z, Winston W, Weiler S, Gyuris J. MAP3K11/GDF15 axis is a critical driver of cancer cachexia. J Cachexia Sarcopenia Muscle 2016; 7:467-82. [PMID: 27239403 PMCID: PMC4863827 DOI: 10.1002/jcsm.12077] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 08/16/2015] [Accepted: 09/10/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cancer associated cachexia affects the majority of cancer patients during the course of the disease and thought to be directly responsible for about a quarter of all cancer deaths. Current evidence suggests that a pro-inflammatory state may be associated with this syndrome although the molecular mechanisms responsible for the development of cachexia are poorly understood. The purpose of this work was the identification of key drivers of cancer cachexia that could provide a potential point of intervention for the treatment and/or prevention of this syndrome. METHODS Genetically engineered and xenograft tumour models were used to dissect the molecular mechanisms driving cancer cachexia. Cytokine profiling from the plasma of cachectic and non-cachectic cancer patients and mouse models was utilized to correlate circulating cytokine levels with the cachexia phenotype. RESULTS Utilizing engineered tumour models we identified MAP3K11/GDF15 pathway activation as a potent inducer of cancer cachexia. Increased expression and high circulating levels of GDF15 acted as a key mediator of this process. In animal models, tumour-produced GDF15 was sufficient to trigger the cachexia phenotype. Elevated GDF15 circulating levels correlated with the onset and progression of cachexia in animal models and in patients with cancer. Inhibition of GDF15 biological activity with a specific antibody reversed body weight loss and restored muscle and fat tissue mass in several cachectic animal models regardless of their complex secreted cytokine profile. CONCLUSIONS The combination of correlative observations, gain of function, and loss of function experiments validated GDF15 as a key driver of cancer cachexia and as a potential therapeutic target for the treatment and/or prevention of this syndrome.
Collapse
Affiliation(s)
- Lorena Lerner
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Julie Tao
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Qing Liu
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | | | - Bin Feng
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Brian Krieger
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Elizabeth Mazsa
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Zakir Siddiquee
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Ruoji Wang
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Lucia Huang
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; Novartis Institutes for BioMedical Research 211 Massachusetts Ave. Cambridge MA 02139 USA
| | - Luhua Shen
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; Moderna Therapeutics 200 Technology Square Cambridge MA 02139 USA
| | - Jie Lin
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; Stealth Peptides Inc.275 Grove Street, Ste.3-107 Newton MA 02466 USA
| | - Antonio Vigano
- McGill Nutrition and Performance Laboratory; (MNUPAL) McGill University Health Centre (MUHC) Montreal Canada
| | - M Isabel Chiu
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; Enumeral Biomedical Corp One Kendall Square Building 400 Cambridge MA 02139 USA
| | - Zhigang Weng
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - William Winston
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; POTENZA Therapeutics 700 Main Street Cambridge MA 02139 USA
| | - Solly Weiler
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Jeno Gyuris
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| |
Collapse
|
4
|
Baratchian M, Davis CA, Shimizu A, Escors D, Bagnéris C, Barrett T, Collins MK. Distinct Activation Mechanisms of NF-κB Regulator Inhibitor of NF-κB Kinase (IKK) by Isoforms of the Cell Death Regulator Cellular FLICE-like Inhibitory Protein (cFLIP). J Biol Chem 2016; 291:7608-20. [PMID: 26865630 PMCID: PMC4817188 DOI: 10.1074/jbc.m116.718122] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Indexed: 11/06/2022] Open
Abstract
The viral FLICE-like inhibitory protein (FLIP) protein from Kaposi sarcoma-associated herpesvirus activates the NF-κB pathway by forming a stable complex with a central region (amino acids 150-272) of the inhibitor of NF-κB kinase (IKK) γ subunits, thereby activating IKK. Cellular FLIP (cFLIP) forms are also known to activate the NF-κB pathway via IKK activation. Here we demonstrate that cFLIPL, cFLIPS, and their proteolytic product p22-FLIP all require the C-terminal region of NEMO/IKKγ (amino acids 272-419) and its ubiquitin binding function for activation of the IKK kinase (or kinase complex), but none form a stable complex with IKKγ. Our results further reveal that cFLIPLrequires the linear ubiquitin chain assembly complex and the kinase TAK1 for activation of the IKK kinase. Similarly, cFLIPSand p22-FLIP also require TAK1 but do not require LUBAC. In contrast, these isoforms are both components of complexes that incorporate Fas-associated death domain and RIP1, which appear essential for kinase activation. This conservation of IKK activation among the cFLIP family using different mechanisms suggests that the mechanism plays a critical role in their function.
Collapse
Affiliation(s)
- Mehdi Baratchian
- From the Medical Research Council Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom, Division of Advanced Therapies, National Institute of Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Herts EN6 3QG, United Kingdom, and
| | - Christopher A Davis
- From the Medical Research Council Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom
| | - Akira Shimizu
- From the Medical Research Council Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom
| | - David Escors
- From the Medical Research Council Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom
| | - Claire Bagnéris
- Institute of Structural and Molecular Biology, School of Biological Sciences, Birkbeck College, Malet Street, London WC1E 7HX, United Kingdom
| | - Tracey Barrett
- Institute of Structural and Molecular Biology, School of Biological Sciences, Birkbeck College, Malet Street, London WC1E 7HX, United Kingdom
| | - Mary K Collins
- From the Medical Research Council Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London WC1E 6BT, United Kingdom, Division of Advanced Therapies, National Institute of Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Herts EN6 3QG, United Kingdom, and
| |
Collapse
|
5
|
Jain P, Lavorgna A, Sehgal M, Gao L, Ginwala R, Sagar D, Harhaj EW, Khan ZK. Myocyte enhancer factor (MEF)-2 plays essential roles in T-cell transformation associated with HTLV-1 infection by stabilizing complex between Tax and CREB. Retrovirology 2015; 12:23. [PMID: 25809782 PMCID: PMC4374383 DOI: 10.1186/s12977-015-0140-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 01/15/2015] [Indexed: 12/17/2022] Open
Abstract
Background The exact molecular mechanisms regarding HTLV-1 Tax-mediated viral gene expression and CD4 T-cell transformation have yet to be fully delineated. Herein, utilizing virus-infected primary CD4+ T cells and the virus-producing cell line, MT-2, we describe the involvement and regulation of Myocyte enhancer factor-2 (specifically MEF-2A) during the course of HTLV-1 infection and associated disease syndrome. Results Inhibition of MEF-2 expression by shRNA and its activity by HDAC9 led to reduced viral replication and T-cell transformation in correlation with a heightened expression of MEF-2 in ATL patients. Mechanistically, MEF-2 was recruited to the viral promoter (LTR, long terminal repeat) in the context of chromatin, and constituted Tax/CREB transcriptional complex via direct binding to the HTLV-1 LTR. Furthermore, an increase in MEF-2 expression was observed upon infection in an extent similar to CREB (known Tax-interacting transcription factor), and HATs (p300, CBP, and p/CAF). Confocal imaging confirmed MEF-2 co-localization with Tax and these proteins were also shown to interact by co-immunoprecipitation. MEF-2 stabilization of Tax/CREB complex was confirmed by a novel promoter-binding assay that highlighted the involvement of NFAT (nuclear factor of activated T cells) in this process via Tax-mediated activation of calcineurin (a calcium-dependent serine-threonine phosphatase). MEF-2-integrated signaling pathways (PI3K/Akt, NF-κB, MAPK, JAK/STAT, and TGF-β) were also activated during HTLV-1 infection of primary CD4+ T cells, possibly regulating MEF-2 activity. Conclusions We demonstrate the involvement of MEF-2 in Tax-mediated LTR activation, viral replication, and T-cell transformation in correlation with its heightened expression in ATL patients through direct binding to DNA within the HTLV-1 LTR. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0140-1) contains supplementary material, which is available to authorized users.
Collapse
|
6
|
Pol JG, Lekbaby B, Redelsperger F, Klamer S, Mandouri Y, Ahodantin J, Bieche I, Lefevre M, Souque P, Charneau P, Gadessaud N, Kremsdorf D, Soussan P. Alternative splicing-regulated protein of hepatitis B virus hacks the TNF-α-stimulated signaling pathways and limits the extent of liver inflammation. FASEB J 2015; 29:1879-89. [PMID: 25630972 DOI: 10.1096/fj.14-258715] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/28/2014] [Indexed: 12/31/2022]
Abstract
Hepatitis B splicing-regulated protein (HBSP) of the hepatitis B virus (HBV) was uncovered a few years ago but its function remains unknown. HBSP expression occurs from a spliced viral transcript that increases during the course of liver disease. This study aimed at characterizing the impact of HBSP on cellular signaling pathways in vitro and on liver pathogenesis in transgenic (Tg) mice. By RT-qPCR array, NF-κB-inducible genes appeared modulated in HepG2 cells transduced with a HBSP-encoding lentivirus. Using luciferase and Western blot assays, we observed a decreased activation of the NF-κB pathway in HBSP-expressing cells following TNF-α treatment, as illustrated by lower levels of phosphorylated IκB-α. Meanwhile, the level of phosphorylated JNK increased together with the sensitivity to apoptosis. The contrasting effects on JNK and IκB-α activation upon TNF-α stimulation matched with a modulated maturation of TGF-β-activated kinase 1 (TAK1) kinase, assessed by 2-dimensional SDS-PAGE. Inhibition of the NF-κB pathway by HBSP was confirmed in the liver of HBSP Tg mice and associated with a significant decrease of chemically induced chronic liver inflammation, as assessed by immunohistochemistry. In conclusion, HBSP contributes to limit hepatic inflammation during chronic liver disease and may favor HBV persistence by evading immune response.
Collapse
Affiliation(s)
- Jonathan G Pol
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Bouchra Lekbaby
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - François Redelsperger
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Sofieke Klamer
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Yassmina Mandouri
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - James Ahodantin
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Ivan Bieche
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Marine Lefevre
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Philippe Souque
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Pierre Charneau
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Noémie Gadessaud
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Dina Kremsdorf
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| | - Patrick Soussan
- *INSERM U845, Pathogenèse des Hépatites Virales B et Immunothérapie, Paris, France; Institut Pasteur, Département de Virologie, Paris, France; Université Paris-Descartes, Centre Hospitalier Universitaire Necker, Paris, France; Laboratoire de Génétique Moléculaire, Faculté des Sciences Pharmaceutiques, Paris, France; Service d'Anatomo-pathologie, Hôpital Tenon, Paris, France; Virologie Moléculaire et Vectorologie, Centre National de la Recherche Scientifique - Unité de Recherche Associée 3015, Institut Pasteur, Paris, France; Plateforme d'Histologie, Laboratoire L-RB126, Paris, France; **Laboratoire de Virologie, Hôpital Tenon, Paris, France; and Université Pierre et Marie Curie, Centre Hospitalier Universitaire Tenon, Paris France
| |
Collapse
|
7
|
Regulation of HTLV-1 tax stability, cellular trafficking and NF-κB activation by the ubiquitin-proteasome pathway. Viruses 2014; 6:3925-43. [PMID: 25341660 PMCID: PMC4213571 DOI: 10.3390/v6103925] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/15/2014] [Accepted: 10/21/2014] [Indexed: 12/22/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is a complex retrovirus that infects CD4+ T cells and causes adult T-cell leukemia/lymphoma (ATLL) in 3%–5% of infected individuals after a long latent period. HTLV-1 Tax is a trans-activating protein that regulates viral gene expression and also modulates cellular signaling pathways to enhance T-cell proliferation and cell survival. The Tax oncoprotein promotes T-cell transformation, in part via constitutive activation of the NF-κB transcription factor; however, the underlying mechanisms remain unknown. Ubiquitination is a type of post-translational modification that occurs in a three-step enzymatic cascade mediated by E1, E2 and E3 enzymes and regulates protein stability as well as signal transduction, protein trafficking and the DNA damage response. Emerging studies indicate that Tax hijacks the ubiquitin machinery to activate ubiquitin-dependent kinases and downstream NF-κB signaling. Tax interacts with the E2 conjugating enzyme Ubc13 and is conjugated on C-terminal lysine residues with lysine 63-linked polyubiquitin chains. Tax K63-linked polyubiquitination may serve as a platform for signaling complexes since this modification is critical for interactions with NEMO and IKK. In addition to NF-κB signaling, mono- and polyubiquitination of Tax also regulate its subcellular trafficking and stability. Here, we review recent advances in the diverse roles of ubiquitin in Tax function and how Tax usurps the ubiquitin-proteasome pathway to promote oncogenesis.
Collapse
|
8
|
Higuchi M, Takahashi M, Tanaka Y, Fujii M. Downregulation of proapoptotic Bim augments IL-2-independent T-cell transformation by human T-cell leukemia virus type-1 Tax. Cancer Med 2014; 3:1605-14. [PMID: 25175936 PMCID: PMC4298387 DOI: 10.1002/cam4.329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 07/26/2014] [Accepted: 08/05/2014] [Indexed: 12/18/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1), an etiological agent of adult T-cell leukemia, immortalizes and transforms primary human T cells in vitro in both an interleukin (IL)-2-dependent and IL-2-independent manner. Expression of the HTLV-1 oncoprotein Tax transforms the growth of the mouse T-cell line CTLL-2 from being IL-2-dependent to IL-2-independent. Withdrawal of IL-2 from normal activated T cells induces apoptosis, which is mediated through the inducible expression of several proapoptotic proteins, including Bim. In this study, we found that Tax protects IL-2-depleted T cells against Bim-induced apoptosis. Withdrawal of IL-2 from CTLL-2 cells induced a prominent increase in the level of Bim protein in CTLL-2 cells, but not in Tax-transformed CTLL-2 cells. This inhibition of Bim in Tax-transformed CTLL-2 cells was mediated by two mechanisms: downregulation of Bim mRNA and posttranscriptional reduction of Bim protein. Transient expression of Tax in CTLL-2 cells also inhibited IL-2 depletion–induced expression of Bim, however, this decrease in Bim protein expression was not due to downregulation of Bim mRNA, thus indicating that Bim mRNA downregulation in Tax-transformed CTLL-2 occurs only after long-term expression of Tax. Transient expression of Tax in CTLL-2 cells also induced Erk activation, however, this was not involved in the reduction of Bim protein. Knockdown of Bim expression in CTLL-2 cells augmented Tax-induced IL-2-independent transformation. HTLV-1 infection of human T cells also reduced their levels of Bim protein, and restoring Bim expression in HTLV-1-infected cells reduced their proliferation by inducing apoptosis. Taken together, these results indicate that Tax-induced downregulation of Bim in HTLV-1-infected T cells promotes their IL-2-independent growth, thereby supporting the persistence of HTLV-1 infection in vivo.
Collapse
Affiliation(s)
- Masaya Higuchi
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | | | | |
Collapse
|
9
|
Liu R, Lin Y, Jia R, Geng Y, Liang C, Tan J, Qiao W. HIV-1 Vpr stimulates NF-κB and AP-1 signaling by activating TAK1. Retrovirology 2014; 11:45. [PMID: 24912525 PMCID: PMC4057933 DOI: 10.1186/1742-4690-11-45] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 05/20/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The Vpr protein of human immunodeficiency virus type 1 (HIV-1) plays an important role in viral replication. It has been reported that Vpr stimulates the nuclear factor-κB (NF-κB) and activator protein 1 (AP-1) signaling pathways, and thereby regulates viral and host cell gene expression. However, the molecular mechanism behind this function of Vpr is not fully understood. RESULTS Here, we have identified transforming growth factor-β-activated kinase 1 (TAK1) as the important upstream signaling molecule that Vpr associates with in order to activate NF-κB and AP-1 signaling. HIV-1 virion-associated Vpr is able to stimulate phosphorylation of TAK1. This activity of Vpr depends on its association with TAK1, since the S79A Vpr mutant lost interaction with TAK1 and was unable to activate TAK1. This association allows Vpr to promote the interaction of TAB3 with TAK1 and increase the polyubiquitination of TAK1, which renders TAK1 phosphorylation. In further support of the key role of TAK1 in this function of Vpr, knockdown of endogenous TAK1 significantly attenuated the ability of Vpr to activate NF-κB and AP-1 as well as the ability to stimulate HIV-1 LTR promoter. CONCLUSIONS HIV-1 Vpr enhances the phosphorylation and polyubiquitination of TAK1, and as a result, activates NF-κB and AP-1 signaling pathways and stimulates HIV-1 LTR promoter.
Collapse
Affiliation(s)
| | | | | | | | | | - Juan Tan
- Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin 300071, China.
| | | |
Collapse
|
10
|
Bidoia C. Human T-lymphotropic virus proteins and post-translational modification pathways. World J Virol 2012; 1:115-30. [PMID: 24175216 PMCID: PMC3782272 DOI: 10.5501/wjv.v1.i4.115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 06/04/2012] [Accepted: 07/13/2012] [Indexed: 02/05/2023] Open
Abstract
Cell life from the cell cycle to the signaling transduction and response to stimuli is finely tuned by protein post-translational modifications (PTMs). PTMs alter the conformation, the stability, the localization, and hence the pattern of interactions of the targeted protein. Cell pathways involve the activation of enzymes, like kinases, ligases and transferases, that, once activated, act on many proteins simultaneously, altering the state of the cell and triggering the processes they are involved in. Viruses enter a balanced system and hijack the cell, exploiting the potential of PTMs either to activate viral encoded proteins or to alter cellular pathways, with the ultimate consequence to perpetuate through their replication. Human T-lymphotropic virus type 1 (HTLV-1) is known to be highly oncogenic and associates with adult T-cell leukemia/lymphoma, HTLV-1-associated myelopathy/tropical spastic paraparesis and other inflammatory pathological conditions. HTLV-1 protein activity is controlled by PTMs and, in turn, viral activity is associated with the modulation of cellular pathways based on PTMs. More knowledge is acquired about the PTMs involved in the activation of its proteins, like Tax, Rex, p12, p13, p30, HTLV-I basic leucine zipper factor and Gag. However, more has to be understood at the biochemical level in order to counteract the associated fatal outcomes. This review will focus on known PTMs that directly modify HTLV-1 components and on enzymes whose activity is modulated by viral proteins.
Collapse
Affiliation(s)
- Carlo Bidoia
- Carlo Bidoia, Centre for Research in Infectious Diseases, School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
11
|
Sakurai H. Targeting of TAK1 in inflammatory disorders and cancer. Trends Pharmacol Sci 2012; 33:522-30. [PMID: 22795313 DOI: 10.1016/j.tips.2012.06.007] [Citation(s) in RCA: 303] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Revised: 06/15/2012] [Accepted: 06/18/2012] [Indexed: 11/18/2022]
Abstract
The transcription factors nuclear factor-κB (NF-κB) and activating protein-1 (AP-1) are critical regulators of stress responses, immunity, inflammation and cancer. A large variety of cellular stimuli utilize these signaling pathways through a common upstream kinase transforming growth factor-β-activated kinase 1 (TAK1). TAK1 was originally identified as a mitogen-activated kinase kinase kinase (MAP3K) activated by transforming growth factor-β (TGF-β); however, it has been characterized as a key regulator in inflammatory and immune signaling pathways. In addition, microbial proteins and components of host cell signaling scramble for the TAK1 complex in innate immunity. This review highlights the recent advances in the activation mechanisms and physiological functions of TAK1. Research targeting TAK1 raises the potential for new therapeutic options for inflammatory disorders, including cancer.
Collapse
Affiliation(s)
- Hiroaki Sakurai
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan.
| |
Collapse
|
12
|
Modulation of autophagy-like processes by tumor viruses. Cells 2012; 1:204-47. [PMID: 24710474 PMCID: PMC3901111 DOI: 10.3390/cells1030204] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 06/13/2012] [Accepted: 06/14/2012] [Indexed: 02/07/2023] Open
Abstract
Autophagy is an intracellular degradation pathway for long-lived proteins and organelles. This process is activated above basal levels upon cell intrinsic or environmental stress and dysregulation of autophagy has been linked to various human diseases, including those caused by viral infection. Many viruses have evolved strategies to directly interfere with autophagy, presumably to facilitate their replication or to escape immune detection. However, in some cases, modulation of autophagy appears to be a consequence of the virus disturbing the cell’s metabolic signaling networks. Here, we summarize recent advances in research at the interface of autophagy and viral infection, paying special attention to strategies that human tumor viruses have evolved.
Collapse
|
13
|
Kaposi's sarcoma associated herpesvirus encoded viral FLICE inhibitory protein K13 activates NF-κB pathway independent of TRAF6, TAK1 and LUBAC. PLoS One 2012; 7:e36601. [PMID: 22590573 PMCID: PMC3348130 DOI: 10.1371/journal.pone.0036601] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 04/11/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Kaposi's sarcoma associated herpesvirus encoded viral FLICE inhibitory protein (vFLIP) K13 activates the NF-κB pathway by binding to the NEMO/IKKγ subunit of the IκB kinase (IKK) complex. However, it has remained enigmatic how K13-NEMO interaction results in the activation of the IKK complex. Recent studies have implicated TRAF6, TAK1 and linear ubiquitin chains assembled by a linear ubiquitin chain assembly complex (LUBAC) consisting of HOIL-1, HOIP and SHARPIN in IKK activation by proinflammatory cytokines. METHODOLOGY/PRINCIPAL FINDINGS Here we demonstrate that K13-induced NF-κB DNA binding and transcriptional activities are not impaired in cells derived from mice with targeted disruption of TRAF6, TAK1 and HOIL-1 genes and in cells derived from mice with chronic proliferative dermatitis (cpdm), which have mutation in the Sharpin gene (Sharpin(cpdm/cpdm)). Furthermore, reconstitution of NEMO-deficient murine embryonic fibroblast cells with NEMO mutants that are incapable of binding to linear ubiquitin chains supported K13-induced NF-κB activity. K13-induced NF-κB activity was not blocked by CYLD, a deubiquitylating enzyme that can cleave linear and Lys63-linked ubiquitin chains. On the other hand, NEMO was required for interaction of K13 with IKK1/IKKα and IKK2/IKKβ, which resulted in their activation by "T Loop" phosphorylation. CONCLUSIONS/SIGNIFICANCE Our results demonstrate that K13 activates the NF-κB pathway by binding to NEMO which results in the recruitment of IKK1/IKKα and IKK2/IKKβ and their subsequent activation by phosphorylation. Thus, K13 activates NF-κB via a mechanism distinct from that utilized by inflammatory cytokines. These results have important implications for the development of therapeutic agents targeting K13-induced NF-κB for the treatment of KSHV-associated malignancies.
Collapse
|
14
|
Qu Z, Xiao G. Human T-cell lymphotropic virus: a model of NF-κB-associated tumorigenesis. Viruses 2011; 3:714-49. [PMID: 21743832 PMCID: PMC3131208 DOI: 10.3390/v3060714] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Revised: 05/13/2011] [Accepted: 06/01/2011] [Indexed: 12/22/2022] Open
Abstract
Human T-cell lymphotropic virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia/lymphoma (ATL), whereas the highly related HTLV-2 is not associated with ATL or other cancers. In addition to ATL leukemogenesis, studies of the HTLV viruses also provide an exceptional model for understanding basic pathogenic mechanisms of virus-host interactions and human oncogenesis. Accumulating evidence suggests that the viral regulatory protein Tax and host inflammatory transcription factor NF-κB are largely responsible for the different pathogenic potentials of HTLV-1 and HTLV-2. Here, we discuss the molecular mechanisms of HTLV-1 oncogenic pathogenesis with a focus on the interplay between the Tax oncoprotein and NF-κB pro-oncogenic signaling. We also outline some of the most intriguing and outstanding questions in the fields of HTLV and NF-κB. Answers to those questions will greatly advance our understanding of ATL leukemogenesis and other NF-κB-associated tumorigenesis and will help us design personalized cancer therapies.
Collapse
Affiliation(s)
- Zhaoxia Qu
- Cancer Institute, Medical Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Gutian Xiao
- Cancer Institute, Medical Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
15
|
Refaat A, Zhou Y, Suzuki S, Takasaki I, Koizumi K, Yamaoka S, Tabuchi Y, Saiki I, Sakurai H. Distinct roles of transforming growth factor-beta-activated kinase 1 (TAK1)-c-Rel and interferon regulatory factor 4 (IRF4) pathways in human T cell lymphotropic virus 1-transformed T helper 17 cells producing interleukin-9. J Biol Chem 2011; 286:21092-9. [PMID: 21498517 DOI: 10.1074/jbc.m110.200907] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Investigation of helper T cell markers in HTLV-1-transformed cell lines demonstrated that HuT-102 has an IL-9-producing Th17 phenotype. We confirmed the vital role of retinoic acid-related orphan receptor C, a Th17 transcription factor, in the expression of IL-17. Interferon regulatory factor 4 (IRF4), a transcription factor overexpressed in all HTLV-1-infected cells, regulated IL-17 and IL-9 concomitantly. We further demonstrated a novel pathway for the regulation of Tax-induced cytokines, IL-9 and IL-6, through TAK1-mediated nuclear accumulation of c-Rel. A microarray analysis for IRF4 knocked down HuT-102 cells showed a significant up-regulation in the set of genes related to Th1, mainly IFN-γ and several transcription factors. T-bet and IRF1, but not STAT1 and IRF9, participated in counteracting the inhibitory effect of IRF4 on the production of IFN-γ. Finally, suppression of both IRF4 and c-Rel resulted in the reduced proliferation. Collectively, these findings indicate that TAK1-c-Rel and IRF4 pathways play distinct roles in the maintenance of IL-9-producing Th17 phenotype of HTLV-1-transformed cells.
Collapse
Affiliation(s)
- Alaa Refaat
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Tareen SU, Emerman M. Human Trim5α has additional activities that are uncoupled from retroviral capsid recognition. Virology 2011; 409:113-20. [PMID: 21035162 PMCID: PMC3051842 DOI: 10.1016/j.virol.2010.09.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 07/05/2010] [Accepted: 09/17/2010] [Indexed: 12/31/2022]
Abstract
Trim5α is a host antiviral protein that recognizes incoming retroviral capsids in the cytoplasm and prevents productive infections. Although present in most mammals, the state of the Trim5 gene is dynamic in that primates have one copy with several splice variants, while rodents and cows have multiple copies. Mouse Trim30 (one of the mouse Trim5α homologs) has been shown to negatively regulate NF-kappaB activation by targeting upstream signaling intermediates TAB2 and TAB3 for degradation. We show that human Trim5α also affects levels of TAB2, resulting in abrogation of TAB2-dependent NF-kappaB activation. Surprisingly, unlike mouse Trim30, human and rhesus Trim5α are able to activate NF-kappaB-driven reporter gene expression in a dose-dependent manner. We show that Trim5α uses distinct domains for the distinct abilities of affecting TAB2 levels, regulating NF-kappaB, and recognizing retroviral capsids. Our results demonstrate functions of Trim5α that are not dependent on recognizing the retroviral capsid.
Collapse
Affiliation(s)
- Semih U. Tareen
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195, USA
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109 USA
| | - Michael Emerman
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109 USA
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109 USA
| |
Collapse
|
17
|
Phosphorylation and polyubiquitination of transforming growth factor beta-activated kinase 1 are necessary for activation of NF-kappaB by the Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor. J Virol 2010; 85:1980-93. [PMID: 21159881 DOI: 10.1128/jvi.01911-10] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) G protein-coupled receptor (vGPCR) protein has been shown to induce several signaling pathways leading to the modulation of host gene expression. The hijacking of these pathways facilitates the viral life cycle and leads to tumorigenesis. In the present work, we show that transforming growth factor β (TGF-β)-activated kinase 1 (TAK1) is an important player in NF-κB activation induced by vGPCR. We observed that the expression of an inactive TAK1 kinase mutant (TAK1M) reduces vGPCR-induced NF-κB nuclear translocation and transcriptional activity. Consequently, the expression of several NF-κB target genes normally induced by vGPCR was blocked by TAK1M expression, including interleukin 8 (IL-8), Gro1, IκBα, COX-2, cIAP2, and Bcl2 genes. Similar results were obtained after downregulation of TAK1 by small interfering RNA (siRNA) technology. The expression of vGPCR recruited TAK1 to the plasma membrane, and vGPCR interacts with TAK1. vGPCR expression also induced TAK1 phosphorylation and lysine 63-linked polyubiquitination, the two markers of the kinase's activation. Finally, inhibition of TAK1 by celastrol inhibited vGPCR-induced NF-κB activation, indicating this natural compound could be used as a potential therapeutic drug against KSHV malignancies involving vGPCR.
Collapse
|
18
|
Association of HTLV Tax proteins with TAK1-binding protein 2 and RelA in calreticulin-containing cytoplasmic structures participates in Tax-mediated NF-κB activation. Virology 2010; 408:39-48. [PMID: 20875659 DOI: 10.1016/j.virol.2010.08.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 06/22/2010] [Accepted: 08/21/2010] [Indexed: 12/20/2022]
Abstract
HTLV-1 is more pathogenic than HTLV-2 despite having a similar genome and closely related transactivating oncoproteins. Both Tax-1 protein from HTLV-1 and Tax-2 from HTLV-2 activate the NF-κB pathway. The mechanisms involved in Tax-1 deregulation of this signalling pathway have been thoroughly investigated, but little is known about regulation by Tax-2. We have compared the interaction of Tax-1 and Tax-2 with two key NF-κB signalling factors: TAK1-binding protein 2 (TAB2), an adaptor involved in the activation of TAK1 kinase, and RelA, the active subunit of the canonical RelA/p50 NF-κB transcription factor. Tax-2 formed stable complexes with both RelA and TAB2. These two NF-κB factors colocalized with Tax proteins in dotted cytoplasmic structures targeted by calreticulin, a multi-process calcium-buffering chaperone. Co-expression of RelA and/or TAB2 markedly increased Tax-mediated NF-κB activation. These findings provide new insights into the role of RelA, TAB2 and Tax in the deregulation of the NF-κB pathway.
Collapse
|
19
|
Shinohara H, Kurosaki T. Comprehending the complex connection between PKCbeta, TAK1, and IKK in BCR signaling. Immunol Rev 2010; 232:300-18. [PMID: 19909372 DOI: 10.1111/j.1600-065x.2009.00836.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The transcription factor nuclear factor-kappaB (NF-kappaB) contributes to many events in the immune system. Characterization of NF-kappaB has facilitated our understanding of immune cell differentiation, survival, proliferation, and effector functions. Intense research continues to elucidate the role of NF-kappaB, which is shared in several receptor signaling pathways, such as Toll-like receptors, the tumor necrosis factor receptor, and antigen receptors. The specificity of cellular responses emanating from stimulation of these receptors is determined by post-translational modification, or 'fine tuning', which regulates spatiotemporal dynamics of downstream signaling. Understanding the fine tuning mechanisms of NF-kappaB activation is crucial for insights into biological regulation and for understanding how cellular signaling pathways are tightly regulated to guide different cell fates. In this review, we focus on recent advances that illuminate the fine tuning mechanisms of NF-kappaB activation by BCR signaling and have increased our comprehension of complex signal systems.
Collapse
Affiliation(s)
- Hisaaki Shinohara
- Laboratory for Lymphocyte Differentiation, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa, Japan.
| | | |
Collapse
|
20
|
Suzuki S, Zhou Y, Refaat A, Takasaki I, Koizumi K, Yamaoka S, Tabuchi Y, Saiki I, Sakurai H. Human T cell lymphotropic virus 1 manipulates interferon regulatory signals by controlling the TAK1-IRF3 and IRF4 pathways. J Biol Chem 2009; 285:4441-6. [PMID: 19955181 DOI: 10.1074/jbc.m109.031476] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
We previously reported that human T cell lymphotropic virus 1 (HTLV-1) Tax oncoprotein constitutively activates transforming growth factor-beta-activated kinase 1 (TAK1). Here, we established Tax-positive HuT-102 cells stably transfected with a short hairpin RNA vector (HuT-shTAK1 cells) and investigated the physiological function of TAK1. Microarray analysis demonstrated that several interferon (IFN)-inducible genes, including chemokines such as CXCL10 and CCL5, were significantly down-regulated in HuT-shTAK1 cells. In contrast, Tax-mediated constitutive activation of nuclear factor-kappaB (NF-kappaB) was intact in HuT-shTAK1 cells. IFN-regulatory factor 3 (IRF3), a critical transcription factor in innate immunity to viral infection, was constitutively activated in a Tax-dependent manner. Activation of IRF3 and IRF3-dependent gene expressions was dependent on TAK1 and TANK-binding kinase 1 (TBK1). On the other hand, IRF4, another member in the IRF family of transcription factors overexpressed in a Tax-independent manner, negatively regulated TAK1-dependent IRF3 transcriptional activity. Together, HTLV-1 manipulates IFN signaling by regulating both positive and negative IRFs.
Collapse
Affiliation(s)
- Shunsuke Suzuki
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Zaidi SFH, Yamamoto T, Refaat A, Ahmed K, Sakurai H, Saiki I, Kondo T, Usmanghani K, Kadowaki M, Sugiyama T. Modulation of activation-induced cytidine deaminase by curcumin in Helicobacter pylori-infected gastric epithelial cells. Helicobacter 2009; 14:588-95. [PMID: 19889077 DOI: 10.1111/j.1523-5378.2009.00724.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Anomalous expression of activation-induced cytidine deaminase (AID) in Helicobacter pylori-infected gastric epithelial cells has been postulated as one of the key mechanisms in the development of gastric cancer. AID is overexpressed in the cells through nuclear factor (NF)-kappaB activation by H. pylori and hence, inhibition of NF-kappaB pathway can downregulate the expression of AID. Curcumin, a spice-derived polyphenol, is known for its anti-inflammatory activity via NF-kappaB inhibition. Therefore, it was hypothesized that curcumin might suppress AID overexpression via NF-kappaB inhibitory activity in H. pylori-infected gastric epithelial cells. MATERIALS AND METHODS MKN-28 or MKN-45 cells and H. pylori strain 193C isolated from gastric cancer patient were used for co-culture experiments. Cells were pretreated with or without nonbactericidal concentrations of curcumin. Apoptosis was determined by DNA fragmentation assay. Enzyme-linked immunosorbent assay was performed to evaluate the anti-adhesion activity of curcumin. Real-time polymerase chain reaction was employed to evaluate the expression of AID mRNA. Immunoblot assay was performed for the analysis of AID, NF-kappaB, inhibitors of NF-kappaB (IkappaB), and IkappaB kinase (IKK) complex regulation with or without curcumin. RESULTS The adhesion of H. pylori to gastric epithelial cells was not inhibited by curcumin pretreatment at nonbactericidal concentrations (< or =10 micromol/L). Pretreatment with nonbactericidal concentration of curcumin downregulated the expression of AID induced by H. pylori. Similarly, NF-kappaB activation inhibitor (SN-50) and proteasome inhibitor (MG-132) also downregulated the mRNA expression of AID. Moreover, curcumin (< or =10 micromol/L) has suppressed H. pylori-induced NF-kappaB activation via inhibition of IKK activation and IkappaB degradation. CONCLUSION Nonbactericidal concentrations of curcumin downregulated H. pylori-induced AID expression in gastric epithelial cells, probably via the inhibition of NF-kappaB pathway. Hence, curcumin can be considered as a potential chemopreventive candidate against H. pylori-related gastric carcinogenesis.
Collapse
Affiliation(s)
- Syed Faisal Haider Zaidi
- Department of Gastroenterology and Hematology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Shin MS, Shinghirunnusorn P, Sugishima Y, Nishimura M, Suzuki S, Koizumi K, Saiki I, Sakurai H. Cross interference with TNF-alpha-induced TAK1 activation via EGFR-mediated p38 phosphorylation of TAK1-binding protein 1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1156-64. [PMID: 19393267 DOI: 10.1016/j.bbamcr.2009.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 03/31/2009] [Accepted: 04/13/2009] [Indexed: 12/17/2022]
Abstract
Transforming growth factor-alpha-activated kinase 1 (TAK1) has been widely recognized as a kinase that regulates multiple intracellular signaling pathways evoked by cytokines and immune receptor activation. We have recently reported that tumor necrosis factor-alpha (TNF-alpha) triggers internalization of epidermal growth factor receptor (EGFR) through a TAK1-p38alpha signaling pathway, which results in a transient suppression of the EGFR. In the present study, we investigated the pathway of intracellular signaling in the opposite direction. Ligand-induced activation of EGFR caused phosphorylation of the TAK1-binding proteins TAB1 and TAB2 in a TAK1-independent manner. EGFR-mediated phosphorylation of TAB1 was completely inhibited by a chemical inhibitor and siRNA of p38alpha. The phosphorylation of TAB1 was occurred at Ser-423 and Thr-431, the residues underlying the p38-mediated feedback inhibition of TAK1. In contrast, phosphorylation of TAB2 was sustained, and largely resistant to p38 inhibition. The inducible phosphorylation of TAB1 interfered with a response of EGF-treated cells to TNF-alpha-induced TAK1 activation, which led to the reduction of NF-kappaB activation. Collectively, these results demonstrated that EGFR activation interfered with TNF-alpha-induced TAK1 activation via p38-mediated phosphorylation of TAB1.
Collapse
Affiliation(s)
- Myoung-Sook Shin
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Boxus M, Twizere JC, Legros S, Dewulf JF, Kettmann R, Willems L. The HTLV-1 Tax interactome. Retrovirology 2008; 5:76. [PMID: 18702816 PMCID: PMC2533353 DOI: 10.1186/1742-4690-5-76] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Accepted: 08/14/2008] [Indexed: 12/22/2022] Open
Abstract
The Tax1 oncoprotein encoded by Human T-lymphotropic virus type I is a major determinant of viral persistence and pathogenesis. Tax1 affects a wide variety of cellular signalling pathways leading to transcriptional activation, proliferation and ultimately transformation. To carry out these functions, Tax1 interacts with and modulates activity of a number of cellular proteins. In this review, we summarize the present knowledge of the Tax1 interactome and propose a rationale for the broad range of cellular proteins identified so far.
Collapse
Affiliation(s)
- Mathieu Boxus
- University Academia Wallonie-Europe, Molecular and Cellular Biology at FUSAGx, Gembloux, Belgium.
| | | | | | | | | | | |
Collapse
|
24
|
Mihaylova VT, Green AM, Khurgel M, Semmes OJ, Kupfer GM. Human T-cell leukemia virus I tax protein sensitizes p53-mutant cells to DNA damage. Cancer Res 2008; 68:4843-52. [PMID: 18559532 PMCID: PMC5939382 DOI: 10.1158/0008-5472.can-07-5070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mutations in p53 are a common cause of resistance of cancers to standard chemotherapy and, thus, treatment failure. Reports have shown that Tax, a human T-cell leukemia virus type I encoded protein that has been associated with genomic instability and perturbation of transcription and cell cycle, sensitizes HeLa cells to UV treatment. The extent to which Tax can sensitize cells and the mechanism by which it exerts its effect are unknown. In this study, we show that Tax sensitizes p53-mutant cells to a broad range of DNA-damaging agents, including mitomycin C, a bifunctional alkylator, etoposide, a topoisomerase II drug, and UV light, but not ionizing radiation, a double-strand break agent, or vinblastine, a tubulin poison. Tax caused hypersensitivity in all p53-deleted cell lines and several, but not all, mutant-expressed p53-containing cell lines, while unexpectedly being protective in p53 wild-type (wt) cells. The effect observed in p53-deleted lines could be reversed for this by transfection of wt p53. We also show that Tax activates a p53-independent proapoptotic program through decreased expression of the retinoblastoma protein and subsequent increased E2F1 expression. The expression of several proapoptotic proteins was also induced by Tax, including Puma and Noxa, culminating in a substantial increase in Bax dimerization. Our results show that Tax can sensitize p53-mutant cells to DNA damage while protecting p53 wt cells, a side benefit that might result in reduced toxicity in normal cells. Such studies hold the promise of a novel adjunctive therapy that could make cancer chemotherapy more effective.
Collapse
Affiliation(s)
- Valia T. Mihaylova
- Department of Microbiology, University of Virginia Health System, Charlottesville, Virginia
- Department of Pediatrics, Yale University, School of Medicine, New Haven, Connecticut
| | - Allison M. Green
- Department of Microbiology, University of Virginia Health System, Charlottesville, Virginia
- Department of Pediatrics, Yale University, School of Medicine, New Haven, Connecticut
| | - Moshe Khurgel
- Department of Microbiology, University of Virginia Health System, Charlottesville, Virginia
| | - Oliver J. Semmes
- Department of Microbiology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Gary M. Kupfer
- Department of Microbiology, University of Virginia Health System, Charlottesville, Virginia
- Department of Pediatrics, University of Virginia Health System, Charlottesville, Virginia
- Department of Pediatrics, Yale University, School of Medicine, New Haven, Connecticut
| |
Collapse
|
25
|
Yu Q, Minoda Y, Yoshida R, Yoshida H, Iha H, Kobayashi T, Yoshimura A, Takaesu G. HTLV-1 Tax-mediated TAK1 activation involves TAB2 adapter protein. Biochem Biophys Res Commun 2007; 365:189-94. [PMID: 17986383 DOI: 10.1016/j.bbrc.2007.10.172] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Accepted: 10/28/2007] [Indexed: 10/22/2022]
Abstract
Human T cell leukemia virus type 1 (HTLV-1) Tax is an oncoprotein that plays a crucial role in the proliferation and transformation of HTLV-1-infected T lymphocytes. It has recently been reported that Tax activates a MAPKKK family, TAK1. However, the molecular mechanism of Tax-mediated TAK1 activation is not well understood. In this report, we investigated the role of TAK1-binding protein 2 (TAB2) in Tax-mediated TAK1 activation. We found that TAB2 physically interacts with Tax and augments Tax-induced NF-kappaB activity. Tax and TAB2 cooperatively activate TAK1 when they are coexpressed. Furthermore, TAK1 activation by Tax requires TAB2 binding as well as ubiquitination of Tax. We also found that the overexpression of TRAF2, 5, or 6 strongly induces Tax ubiquitination. These results suggest that TAB2 may be critically involved in Tax-mediated activation of TAK1 and that NF-kappaB-activating TRAF family proteins are potential cellular E3 ubiquitin ligases toward Tax.
Collapse
Affiliation(s)
- Qingsheng Yu
- Division of Molecular and Cellular Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
The human T-cell leukemia virus type 1 Tax oncoprotein requires the ubiquitin-conjugating enzyme Ubc13 for NF-kappaB activation. J Virol 2007; 81:13735-42. [PMID: 17942533 DOI: 10.1128/jvi.01790-07] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Ubiquitination of the human T-cell leukemia virus 1 Tax oncoprotein provides an important regulatory mechanism that promotes the Tax-mediated activation of NF-kappaB. However, the type of polyubiquitin chain linkages and the host factors that are required for Tax ubiquitination have not been identified. Here, we demonstrate that Tax polyubiquitin chains are composed predominantly of lysine 63-linked chains. Furthermore, the ubiquitination of Tax is critically dependent on the E2 ubiquitin-conjugating enzyme Ubc13. Tax interacts with Ubc13, and small interfering RNA-mediated knockdown of Ubc13 expression abrogates Tax ubiquitination and the activation of NF-kappaB. Mouse fibroblasts lacking Ubc13 exhibit impaired Tax activation of NF-kappaB despite normal tumor necrosis factor- and interleukin-1-mediated NF-kappaB activation. Finally, the interaction of Tax with NEMO is disrupted in the absence of Tax ubiquitination and Ubc13 expression, suggesting that Tax ubiquitination is critical for NEMO binding. Collectively, our results reveal that Ubc13 is essential for Tax ubiquitination, its interaction with NEMO, and Tax-mediated NF-kappaB activation.
Collapse
|