1
|
Sato N, Goyama S, Chang YH, Miyawaki M, Fujino T, Koide S, Denda T, Liu X, Ueda K, Yamamoto K, Asada S, Takeda R, Yonezawa T, Tanaka Y, Honda H, Ota Y, Shibata T, Sekiya M, Isobe T, Lamagna C, Masuda E, Iwama A, Shimano H, Inoue JI, Miyake K, Kitamura T. Clonal hematopoiesis-related mutant ASXL1 promotes atherosclerosis in mice via dysregulated innate immunity. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1568-1583. [PMID: 39653824 DOI: 10.1038/s44161-024-00579-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024]
Abstract
Certain somatic mutations provide a fitness advantage to hematopoietic stem cells and lead to clonal expansion of mutant blood cells, known as clonal hematopoiesis (CH). Among the most common CH mutations, ASXL1 mutations pose the highest risk for cardiovascular diseases (CVDs), yet the mechanisms by which they contribute to CVDs are unclear. Here we show that hematopoietic cells harboring C-terminally truncated ASXL1 mutant (ASXL1-MT) accelerate the development of atherosclerosis in Ldlr-/- mice. Transcriptome analyses of plaque cells showed that monocytes and macrophages expressing ASXL1-MT exhibit inflammatory signatures. Mechanistically, we demonstrate that wild-type ASXL1 has an unexpected non-epigenetic role by suppressing innate immune signaling through the inhibition of IRAK1-TAK1 interaction in the cytoplasm. This regulatory function is lost in ASXL1-MT, resulting in NF-κB activation. Inhibition of IRAK1/4 alleviated atherosclerosis driven by ASXL1-MT and decreased inflammatory monocytes. The present work provides a mechanistic and cellular explanation linking ASXL1 mutations, CH and CVDs.
Collapse
Affiliation(s)
- Naru Sato
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-Ku, Japan
| | - Susumu Goyama
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Yu-Hsuan Chang
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-Ku, Japan
| | - Masashi Miyawaki
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-Ku, Japan
| | - Takeshi Fujino
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shuhei Koide
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tamami Denda
- Department of Pathology, The Institute of Medical Science Research Hospital, University of Tokyo, Tokyo, Japan
| | - Xiaoxiao Liu
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Koji Ueda
- Project for Personalized Cancer Medicine, Cancer Precision Medicine Centre, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Keita Yamamoto
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Shuhei Asada
- The Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Reina Takeda
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Taishi Yonezawa
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yosuke Tanaka
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroaki Honda
- The Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Yasunori Ota
- Department of Pathology, The Institute of Medical Science Research Hospital, University of Tokyo, Tokyo, Japan
| | - Takuma Shibata
- Department of Microbiology and Immunology, Division of Infectious Genetics, University of Tokyo, Tokyo, Japan
| | - Motohiro Sekiya
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tomoya Isobe
- Department of Hematology, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | | | - Esteban Masuda
- Rigel Pharmaceuticals, Inc., South San Francisco, CA, USA
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Jun-Ichiro Inoue
- Research Platform Office, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kensuke Miyake
- Department of Microbiology and Immunology, Division of Infectious Genetics, University of Tokyo, Tokyo, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-Ku, Japan.
- Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan.
| |
Collapse
|
2
|
Cao X, Tan J, Zheng R, Wang F, Zhou L, Yi J, Yuan R, Dai Q, Song L, Dai A. Targeting necroptosis: a promising avenue for respiratory disease treatment. Cell Commun Signal 2024; 22:418. [PMID: 39192326 DOI: 10.1186/s12964-024-01804-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
Respiratory diseases are a growing concern in public health because of their potential to endanger the global community. Cell death contributes critically to the pathophysiology of respiratory diseases. Recent evidence indicates that necroptosis, a unique form of programmed cell death (PCD), plays a vital role in the molecular mechanisms underlying respiratory diseases, distinguishing it from apoptosis and conventional necrosis. Necroptosis is a type of inflammatory cell death governed by receptor-interacting serine/threonine protein kinase 1 (RIPK1), RIPK3, and mixed-lineage kinase domain-like protein (MLKL), resulting in the release of intracellular contents and inflammatory factors capable of initiating an inflammatory response in adjacent tissues. These necroinflammatory conditions can result in significant organ dysfunction and long-lasting tissue damage within the lungs. Despite evidence linking necroptosis to various respiratory diseases, there are currently no specific alternative treatments that target this mechanism. This review provides a comprehensive overview of the most recent advancements in understanding the significance and mechanisms of necroptosis. Specifically, this review emphasizes the intricate association between necroptosis and respiratory diseases, highlighting the potential use of necroptosis as an innovative therapeutic approach for treating these conditions.
Collapse
Affiliation(s)
- Xianya Cao
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Junlan Tan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Runxiu Zheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Feiying Wang
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Lingling Zhou
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Jian Yi
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Rong Yuan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Qin Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Lan Song
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
3
|
Connor MG, Hamon MA. Advances in regulation of homeostasis through chromatin modifications by airway commensals. Curr Opin Microbiol 2024; 80:102505. [PMID: 38936013 DOI: 10.1016/j.mib.2024.102505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024]
Abstract
Commensal bacteria are residents of the human airway where they interact with both colonizing pathogens and host respiratory epithelial cells of this mucosal surface. It is here that commensals exert their influence through host signaling cascades, host transcriptional responses and host immunity, all of which are rooted in chromatin remodeling and histone modifications. Recent studies show that airway commensals impact host chromatin, but compared the what is known for gut commensals, the field remains in its infancy. The mechanisms by which airway commensals regulate respiratory health and homeostasis through chromatin modifications is of increasing interest, specifically since their displacement precedes the increased potential for respiratory disease. Herein we will discuss recent advances and intriguing avenues of future work aimed at deciphering how airway commensals protect and influence respiratory health.
Collapse
Affiliation(s)
- Michael G Connor
- Institut Pasteur, Université de Paris Cité, Unité Chromatine et Infection, F-75015 Paris, France.
| | - Melanie A Hamon
- Institut Pasteur, Université de Paris Cité, Unité Chromatine et Infection, F-75015 Paris, France.
| |
Collapse
|
4
|
Rogers NM, Zammit N, Nguyen-Ngo D, Souilmi Y, Minhas N, Meijles DN, Self E, Walters SN, Warren J, Cultrone D, El-Rashid M, Li J, Chtanova T, O'Connell PJ, Grey ST. The impact of the cytoplasmic ubiquitin ligase TNFAIP3 gene variation on transcription factor NF-κB activation in acute kidney injury. Kidney Int 2023; 103:1105-1119. [PMID: 37097268 DOI: 10.1016/j.kint.2023.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/08/2023] [Accepted: 02/23/2023] [Indexed: 04/26/2023]
Abstract
Nuclear factor κB (NF-κB) activation is a deleterious molecular mechanism that drives acute kidney injury (AKI) and manifests in transplanted kidneys as delayed graft function. The TNFAIP3 gene encodes A20, a cytoplasmic ubiquitin ligase and a master negative regulator of the NF- κB signaling pathway. Common population-specific TNFAIP3 coding variants that reduce A20's enzyme function and increase NF- κB activation have been linked to heightened protective immunity and autoimmune disease, but have not been investigated in AKI. Here, we functionally identified a series of unique human TNFAIP3 coding variants linked to the autoimmune genome-wide association studies single nucleotide polymorphisms of F127C; namely F127C;R22Q, F127C;G281E, F127C;W448C and F127C;N449K that reduce A20's anti-inflammatory function in an NF- κB reporter assay. To investigate the impact of TNFAIP3 hypomorphic coding variants in AKI we tested a mouse Tnfaip3 hypomorph in a model of ischemia reperfusion injury (IRI). The mouse Tnfaip3 coding variant I325N increases NF- κB activation without overt inflammatory disease, providing an immune boost as I325N mice exhibit enhanced innate immunity to a bacterial challenge. Surprisingly, despite exhibiting increased intra-kidney NF- κB activation with inflammation in IRI, the kidney of I325N mice was protected. The I325N variant influenced the outcome of IRI by changing the dynamic expression of multiple cytoprotective mechanisms, particularly by increasing NF- κB-dependent anti-apoptotic factors BCL-2, BCL-XL, c-FLIP and A20, altering the active redox state of the kidney with a reduction of superoxide levels and the enzyme super oxide dismutase-1, and enhancing cellular protective mechanisms including increased Foxp3+ T cells. Thus, TNFAIP3 gene variants represent a kidney and population-specific molecular factor that can dictate the course of IRI.
Collapse
Affiliation(s)
- Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Renal and Transplant Medicine Unit, Westmead Hospital, Westmead, New South Wales, Australia; Westmead Clinical School, University of Sydney, New South Wales, Australia
| | - Nathan Zammit
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Danny Nguyen-Ngo
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Yassine Souilmi
- Australian Centre for Ancient DNA, School of Biological Sciences, University of Adelaide, South Australia, Australia; Environment Institute, Faculty of Sciences, University of Adelaide, South Australia, Australia
| | - Nikita Minhas
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - Eleanor Self
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Stacey N Walters
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Joanna Warren
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Daniele Cultrone
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Maryam El-Rashid
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Tatyana Chtanova
- Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Innate and Tumour Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Philip J O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia; Renal and Transplant Medicine Unit, Westmead Hospital, Westmead, New South Wales, Australia; Westmead Clinical School, University of Sydney, New South Wales, Australia
| | - Shane T Grey
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; Translational Research Pillar, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
5
|
Rohwedder I, Wackerbarth LM, Heinig K, Ballweg A, Altstätter J, Ripphahn M, Nussbaum C, Salvermoser M, Bierschenk S, Straub T, Gunzer M, Schmidt-Supprian M, Kolben T, Schulz C, Ma A, Walzog B, Heinig M, Sperandio M. A20 and the noncanonical NF-κB pathway are key regulators of neutrophil recruitment during fetal ontogeny. JCI Insight 2023; 8:155968. [PMID: 36633909 PMCID: PMC9977499 DOI: 10.1172/jci.insight.155968] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/11/2023] [Indexed: 01/13/2023] Open
Abstract
Newborns are at high risk of developing neonatal sepsis, particularly if born prematurely. This has been linked to divergent requirements the immune system has to fulfill during intrauterine compared with extrauterine life. By transcriptomic analysis of fetal and adult neutrophils, we shed new light on the molecular mechanisms of neutrophil maturation and functional adaption during fetal ontogeny. We identified an accumulation of differentially regulated genes within the noncanonical NF-κB signaling pathway accompanied by constitutive nuclear localization of RelB and increased surface expression of TNF receptor type II in fetal neutrophils, as well as elevated levels of lymphotoxin α in fetal serum. Furthermore, we found strong upregulation of the negative inflammatory regulator A20 (Tnfaip3) in fetal neutrophils, which was accompanied by pronounced downregulation of the canonical NF-κB pathway. Functionally, overexpressing A20 in Hoxb8 cells led to reduced adhesion of these neutrophil-like cells in a flow chamber system. Conversely, mice with a neutrophil-specific A20 deletion displayed increased inflammation in vivo. Taken together, we have uncovered constitutive activation of the noncanonical NF-κB pathway with concomitant upregulation of A20 in fetal neutrophils. This offers perfect adaption of neutrophil function during intrauterine fetal life but also restricts appropriate immune responses particularly in prematurely born infants.
Collapse
Affiliation(s)
- Ina Rohwedder
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Lou Martha Wackerbarth
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Kristina Heinig
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Annamaria Ballweg
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Johannes Altstätter
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Myriam Ripphahn
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Claudia Nussbaum
- Division of Neonatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU Munich, Munich, Germany
| | - Melanie Salvermoser
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Susanne Bierschenk
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Tobias Straub
- Core Facility Bioinformatics, Biomedical Center Munich, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University of Duisburg-Essen, Essen, Germany.,Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | | | | | - Christian Schulz
- Medical Clinic I, University Hospital, LMU Munich, Munich, Germany
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Barbara Walzog
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| | - Matthias Heinig
- Institute of Computational Biology, Helmholtz Munich, Munich, Germany.,Department of Informatics, TU Munich, Munich, Germany
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and Pathophysiology, Walter-Brendel-Center of Experimental Medicine, Biomedical Center Munich, LMU Munich, Planegg-Martinsried, Germany
| |
Collapse
|
6
|
McLemore AF, Hou HA, Meyer BS, Lam NB, Ward GA, Aldrich AL, Rodrigues MA, Vedder A, Zhang L, Padron E, Vincelette ND, Sallman DA, Abdel-Wahab O, List AF, McGraw KL. Somatic gene mutations expose cytoplasmic DNA to co-opt the cGAS-STING-NLRP3 axis in Myelodysplastic syndromes. JCI Insight 2022; 7:159430. [PMID: 35788117 PMCID: PMC9462508 DOI: 10.1172/jci.insight.159430] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/01/2022] [Indexed: 11/17/2022] Open
Abstract
NLRP3 inflammasome and IFN-stimulated gene (ISG) induction are key biological drivers of ineffective hematopoiesis and inflammation in myelodysplastic syndromes (MDSs). Gene mutations involving mRNA splicing and epigenetic regulatory pathways induce inflammasome activation and myeloid lineage skewing in MDSs through undefined mechanisms. Using immortalized murine hematopoietic stem and progenitor cells harboring these somatic gene mutations and primary MDS BM specimens, we showed accumulation of unresolved R-loops and micronuclei with concurrent activation of the cytosolic sensor cyclic GMP-AMP synthase. Cyclic GMP-AMP synthase/stimulator of IFN genes (cGAS/STING) signaling caused ISG induction, NLRP3 inflammasome activation, and maturation of the effector protease caspase-1. Deregulation of RNA polymerase III drove cytosolic R-loop generation, which upon inhibition, extinguished ISG and inflammasome response. Mechanistically, caspase-1 degraded the master erythroid transcription factor, GATA binding protein 1, provoking anemia and myeloid lineage bias that was reversed by cGAS inhibition in vitro and in Tet2–/– hematopoietic stem and progenitor cell–transplanted mice. Together, these data identified a mechanism by which functionally distinct mutations converged upon the cGAS/STING/NLRP3 axis in MDS, directing ISG induction, pyroptosis, and myeloid lineage skewing.
Collapse
Affiliation(s)
- Amy F McLemore
- Department of Malignant Hematology, Moffitt Cancer Center & Research institute, Tampa, United States of America
| | - Hsin-An Hou
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Benjamin S Meyer
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States of America
| | - Nghi B Lam
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States of America
| | - Grace A Ward
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States of America
| | - Amy L Aldrich
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States of America
| | | | - Alexis Vedder
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States of America
| | - Ling Zhang
- Department of Hemapathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States of America
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States of America
| | - Nicole D Vincelette
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States of America
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, United States of America
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, United States of America
| | - Alan F List
- Precision Biosciences, Precision Biosciences, Durham, United States of America
| | - Kathy L McGraw
- Center for Cancer Research, National Cancer Institute, Bethesda, United States of America
| |
Collapse
|
7
|
A20 undermines alternative NF-κB activity and expression of anti-apoptotic genes in Helicobacter pylori infection. Cell Mol Life Sci 2022; 79:102. [PMID: 35089437 PMCID: PMC8799570 DOI: 10.1007/s00018-022-04139-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/16/2021] [Accepted: 01/08/2022] [Indexed: 12/16/2022]
Abstract
A hallmark of infection by the pathogen Helicobacter pylori, which colonizes the human gastric epithelium, is the simultaneous activation of the classical and alternative nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways, underlying inflammation and cell survival. Here, we report that the classical NF-κB target gene product A20 contributes to the negative regulation of alternative NF-κB signaling in gastric epithelial cells infected by H. pylori. Mechanistically, the de novo synthesized A20 protein interacts with tumor necrosis factor receptor-associated factor-interacting protein with forkhead-associated domain (TIFA) and thereby interferes with the association of TIFA with the NIK regulatory complex. We also show that alternative NF-κB activity contributes to the up-regulation of anti-apoptotic genes, such as baculoviral IAP repeat containing 2 (BIRC2), BIRC3 and B-cell lymphoma 2-related protein A1 (BCL2A1) in gastric epithelial cells. Furthermore, the observed over-expression of RelB in human gastric biopsies with type B gastritis and RelB-dependent suppression of apoptotic cell death emphasize an important role of the alternative NF-κB pathway in H. pylori infection.
Collapse
|
8
|
Chronic exposure to TNF reprograms cell signaling pathways in fibroblast-like synoviocytes by establishing long-term inflammatory memory. Sci Rep 2020; 10:20297. [PMID: 33219307 PMCID: PMC7679373 DOI: 10.1038/s41598-020-77380-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/28/2020] [Indexed: 12/31/2022] Open
Abstract
Fibroblast-like synoviocytes (FLS) play a critical role in the pathogenesis of rheumatoid arthritis (RA). Chronic inflammation induces transcriptomic and epigenetic modifications that imparts a persistent catabolic phenotype to the FLS, despite their dissociation from the inflammatory environment. We analyzed high throughput gene expression and chromatin accessibility data from human and mouse FLS from our and other studies available on public repositories, with the goal of identifying the persistently reprogrammed signaling pathways driven by chronic inflammation. We found that the gene expression changes induced by short-term tumor necrosis factor-alpha (TNF) treatment were largely sustained in the FLS exposed to chronic inflammation. These changes that included both activation and repression of gene expression, were accompanied by the remodeling of chromatin accessibility. The sustained activated genes (SAGs) included established pro-inflammatory signaling components known to act at multiple levels of NF-kappaB, STAT and AP-1 signaling cascades. Interestingly, the sustained repressed genes (SRGs) included critical mediators and targets of the BMP signaling pathway. We thus identified sustained repression of BMP signaling as a unique constituent of the long-term inflammatory memory induced by chronic inflammation. We postulate that simultaneous targeting of these activated and repressed signaling pathways may be necessary to combat RA persistence.
Collapse
|
9
|
Angelini DF, De Angelis F, Vacca V, Piras E, Parisi C, Nutini M, Spalloni A, Pagano F, Longone P, Battistini L, Pavone F, Marinelli S. Very Early Involvement of Innate Immunity in Peripheral Nerve Degeneration in SOD1-G93A Mice. Front Immunol 2020; 11:575792. [PMID: 33329541 PMCID: PMC7714949 DOI: 10.3389/fimmu.2020.575792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Recent preclinical and clinical evidence suggest that immune system has a role in the progression and prognosis of Amyotrophic Lateral Sclerosis (ALS), but the identification of a clear mechanism and immune players remains to be elucidated. Here, we have investigated, in 30 and 60 days (presymptomatic) and 120 days (symptomatic) old SOD1-G93A mice, systemic, peripheral, and central innate and adaptive immune and inflammatory response, correlating it with the progression of the neurodegeneration in neuromuscular junction, sciatic nerves, and spinal cord. Surprisingly, we found a very initial (45-60 days) presence of IgG in sciatic nerves together with a gradual enhancement of A20/TNFAIP3 (protein controlling NF-κB signalling) and a concomitantly significant increase and activation of circulating mast cells (MCs) as well as MCs and macrophages in sciatic nerve and an enhancement of IL-6 and IL-10. This immunological frame coincided with a myelin aggregation. The 30-60 days old SOD1-G93A mice didn't show real elements of neuroinflammation and neurodegeneration in spinal cord. In 120 days old mice macrophages and monocytes are widely diffused in sciatic nerves, peripheral neurodegeneration reaches the tip, high circulating levels of TNFα and IL-2 were found and spinal cord exhibits clear signs of neural damage and infiltrating immune cells. Our results underpin a clear immunological disorder at the origin of ALS axonopathy, in which MCs are involved in the initiation and sustaining of inflammatory events. These data cannot be considered a mere epiphenomenon of motor neuron degeneration and reveal new potential selective immune targets in ALS therapy.
Collapse
Affiliation(s)
| | - Federica De Angelis
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Valentina Vacca
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Eleonora Piras
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Chiara Parisi
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Michele Nutini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Alida Spalloni
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Francesca Pagano
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | | | - Luca Battistini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Flaminia Pavone
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Sara Marinelli
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| |
Collapse
|
10
|
Datar GK, Goodell MA. Where There’s Smoke, There’s Fire: Inflammation Drives MDS. Trends Immunol 2020; 41:558-560. [DOI: 10.1016/j.it.2020.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 05/23/2020] [Indexed: 12/16/2022]
|
11
|
|
12
|
Muto T, Walker CS, Choi K, Hueneman K, Smith MA, Gul Z, Garcia-Manero G, Ma A, Zheng Y, Starczynowski DT. Adaptive response to inflammation contributes to sustained myelopoiesis and confers a competitive advantage in myelodysplastic syndrome HSCs. Nat Immunol 2020; 21:535-545. [PMID: 32313245 DOI: 10.1038/s41590-020-0663-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/17/2020] [Indexed: 02/06/2023]
Abstract
Despite evidence of chronic inflammation in myelodysplastic syndrome (MDS) and cell-intrinsic dysregulation of Toll-like receptor (TLR) signaling in MDS hematopoietic stem and progenitor cells (HSPCs), the mechanisms responsible for the competitive advantage of MDS HSPCs in an inflammatory milieu over normal HSPCs remain poorly defined. Here, we found that chronic inflammation was a determinant for the competitive advantage of MDS HSPCs and for disease progression. The cell-intrinsic response of MDS HSPCs, which involves signaling through the noncanonical NF-κB pathway, protected these cells from chronic inflammation as compared to normal HSPCs. In response to inflammation, MDS HSPCs switched from canonical to noncanonical NF-κB signaling, a process that was dependent on TLR-TRAF6-mediated activation of A20. The competitive advantage of TLR-TRAF6-primed HSPCs could be restored by deletion of A20 or inhibition of the noncanonical NF-κB pathway. These findings uncover the mechanistic basis for the clonal dominance of MDS HSPCs and indicate that interfering with noncanonical NF-κB signaling could prevent MDS progression.
Collapse
Affiliation(s)
- Tomoya Muto
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Callum S Walker
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kathleen Hueneman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Molly A Smith
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zartash Gul
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | | | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
13
|
Martín-Vicente M, González-Sanz R, Cuesta I, Monzón S, Resino S, Martínez I. Downregulation of A20 Expression Increases the Immune Response and Apoptosis and Reduces Virus Production in Cells Infected by the Human Respiratory Syncytial Virus. Vaccines (Basel) 2020; 8:vaccines8010100. [PMID: 32102364 PMCID: PMC7157707 DOI: 10.3390/vaccines8010100] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 01/20/2023] Open
Abstract
Human respiratory syncytial virus (HRSV) causes severe lower respiratory tract infections in infants, the elderly, and immunocompromised adults. Regulation of the immune response against HRSV is crucial to limiting virus replication and immunopathology. The A20/TNFAIP3 protein is a negative regulator of nuclear factor kappa B (NF-κB) and interferon regulatory factors 3/7 (IRF3/7), which are key transcription factors involved in the inflammatory/antiviral response of epithelial cells to virus infection. Here, we investigated the impact of A20 downregulation or knockout on HRSV growth and the induction of the immune response in those cells. Cellular infections in which the expression of A20 was silenced by siRNAs or eliminated by gene knockout showed increased inflammatory/antiviral response and reduced virus production. Similar results were obtained when the expression of A20-interacting proteins, such as TAX1BP1 and ABIN1, was silenced. Additionally, downregulation of A20, TAX1BP1, and ABIN1 increased cell apoptosis in HRSV-infected cells. These results show that the downregulation of A20 expression might contribute in the control of HRSV infections by potentiating the early innate immune response and increasing apoptosis in infected cells.
Collapse
Affiliation(s)
- María Martín-Vicente
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-V.); (R.G.-S.); (S.R.)
| | - Rubén González-Sanz
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-V.); (R.G.-S.); (S.R.)
| | - Isabel Cuesta
- Unidad de Bioinformática, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (I.C.); (S.M.)
| | - Sara Monzón
- Unidad de Bioinformática, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (I.C.); (S.M.)
| | - Salvador Resino
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-V.); (R.G.-S.); (S.R.)
| | - Isidoro Martínez
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-V.); (R.G.-S.); (S.R.)
- Correspondence: ; Tel.: +34-91-8223272; Fax: +34-91-5097919
| |
Collapse
|
14
|
Feoktistova M, Makarov R, Brenji S, Schneider AT, Hooiveld GJ, Luedde T, Leverkus M, Yazdi AS, Panayotova-Dimitrova D. A20 Promotes Ripoptosome Formation and TNF-Induced Apoptosis via cIAPs Regulation and NIK Stabilization in Keratinocytes. Cells 2020; 9:E351. [PMID: 32028675 PMCID: PMC7072579 DOI: 10.3390/cells9020351] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/29/2020] [Accepted: 02/01/2020] [Indexed: 12/30/2022] Open
Abstract
The ubiquitin-editing protein A20 (TNFAIP3) is a known key player in the regulation of immune responses in many organs. Genome-wide associated studies (GWASs) have linked A20 with a number of inflammatory and autoimmune disorders, including psoriasis. Here, we identified a previously unrecognized role of A20 as a pro-apoptotic factor in TNF-induced cell death in keratinocytes. This function of A20 is mediated via the NF-κB-dependent alteration of cIAP1/2 expression. The changes in cIAP1/2 protein levels promote NIK stabilization and subsequent activation of noncanonical NF-κB signaling. Upregulation of TRAF1 expression triggered by the noncanonical NF-κB signaling further enhances the NIK stabilization in an autocrine manner. Finally, stabilized NIK promotes the formation of the ripoptosome and the execution of cell death. Thus, our data demonstrate that A20 controls the execution of TNF-induced cell death on multiple levels in keratinocytes. This signaling mechanism might have important implications for the development of new therapeutic strategies for the treatment of A20-associated skin diseases.
Collapse
Affiliation(s)
- Maria Feoktistova
- Department of Dermatology and Allergology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (M.F.); (R.M.); (S.B.); (A.S.Y.)
| | - Roman Makarov
- Department of Dermatology and Allergology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (M.F.); (R.M.); (S.B.); (A.S.Y.)
| | - Sihem Brenji
- Department of Dermatology and Allergology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (M.F.); (R.M.); (S.B.); (A.S.Y.)
| | - Anne T. Schneider
- Department of Medicine III, Department of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (A.T.S.); (T.L.)
| | - Guido J. Hooiveld
- Nutrition, Metabolism & Genomics Group, Division of Human Nutrition & Health, Wageningen University, 6700 AA Wageningen; The Netherlands;
| | - Tom Luedde
- Department of Medicine III, Department of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (A.T.S.); (T.L.)
| | - Martin Leverkus
- Department of Dermatology and Allergology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (M.F.); (R.M.); (S.B.); (A.S.Y.)
| | - Amir S. Yazdi
- Department of Dermatology and Allergology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (M.F.); (R.M.); (S.B.); (A.S.Y.)
| | - Diana Panayotova-Dimitrova
- Department of Dermatology and Allergology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (M.F.); (R.M.); (S.B.); (A.S.Y.)
| |
Collapse
|
15
|
Yang X, Cheng H, Chen J, Wang R, Saleh A, Si H, Lee S, Guven-Maiorov E, Keskin O, Gursoy A, Nussinov R, Fang J, Van Waes C, Chen Z. Head and Neck Cancers Promote an Inflammatory Transcriptome through Coactivation of Classic and Alternative NF-κB Pathways. Cancer Immunol Res 2019; 7:1760-1774. [PMID: 31624067 PMCID: PMC6941750 DOI: 10.1158/2326-6066.cir-18-0832] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 04/22/2019] [Accepted: 09/03/2019] [Indexed: 01/01/2023]
Abstract
Head and neck squamous cell carcinomas (HNSCC) promote inflammation in the tumor microenvironment through aberrant NF-κB activation, but the genomic alterations and pathway networks that modulate NF-κB signaling have not been fully dissected. Here, we analyzed genome and transcriptome alterations of 279 HNSCC specimens from The Cancer Genome Atlas (TCGA) cohort and identified 61 genes involved in NF-κB and inflammatory pathways. The top 30 altered genes were distributed across 96% of HNSCC samples, and their expression was often correlated with genomic copy-number alterations (CNA). Ten of the amplified genes were associated with human papilloma virus (HPV) status. We sequenced 15 HPV- and 11 HPV+ human HNSCC cell lines, and three oral mucosa keratinocyte lines, and supervised clustering revealed that 28 of 61 genes exhibit altered expression patterns concordant with HNSCC tissues and distinct signatures related to their HPV status. RNAi screening using an NF-κB reporter line identified 16 genes that are induced by TNFα or Lymphotoxin-β (LTβ) and implicated in the classic and/or alternative NF-κB pathways. Knockdown of TNFR, LTBR, or selected downstream signaling components established cross-talk between the classic and alternative NF-κB pathways. TNFα and LTβ induced differential gene expression involving the NF-κB, IFNγ, and STAT pathways, inflammatory cytokines, and metastasis-related genes. Improved survival was observed in HNSCC patients with elevated gene expression in T-cell activation, immune checkpoints, and IFNγ and STAT pathways. These gene signatures of NF-κB activation, which modulate inflammation and responses to the immune therapy, could serve as potential biomarkers in future clinical trials.
Collapse
Affiliation(s)
- Xinping Yang
- Tumor Biology Section and Clinical Genomics Unit, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Hui Cheng
- Tumor Biology Section and Clinical Genomics Unit, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Jianhong Chen
- Tumor Biology Section and Clinical Genomics Unit, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Ru Wang
- Tumor Biology Section and Clinical Genomics Unit, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Anthony Saleh
- Tumor Biology Section and Clinical Genomics Unit, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Han Si
- Tumor Biology Section and Clinical Genomics Unit, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Steven Lee
- Tumor Biology Section and Clinical Genomics Unit, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| | - Emine Guven-Maiorov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
- Department of Chemical and Biological Engineering, College of Engineering, Koc University, Istanbul, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, College of Engineering, Koc University, Istanbul, Turkey
- Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Attila Gursoy
- Department of Computer Engineering, College of Engineering, Koc University, Istanbul, Turkey
- Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jugao Fang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Carter Van Waes
- Tumor Biology Section and Clinical Genomics Unit, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland.
| | - Zhong Chen
- Tumor Biology Section and Clinical Genomics Unit, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
16
|
Sánchez-Martín D, Otsuka A, Kabashima K, Ha T, Wang D, Qian X, Lowy DR, Tosato G. Effects of DLC1 Deficiency on Endothelial Cell Contact Growth Inhibition and Angiosarcoma Progression. J Natl Cancer Inst 2019; 110:390-399. [PMID: 29202196 DOI: 10.1093/jnci/djx219] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/18/2017] [Indexed: 01/04/2023] Open
Abstract
Background Deleted in Liver Cancer 1 (DLC1) is a tumor suppressor gene frequently deleted in cancer. However, DLC1 is not known to be deleted in angiosarcoma, an aggressive malignancy of endothelial cell derivation. Additionally, the physiologic functions of DLC1 protein in endothelial cells are poorly defined. Methods We investigated the effects of shRNA-induced DLC1 depletion in endothelial cells. Cell growth was measured by 3H thymidine incorporation, IncuCyte imaging, and population doublings; cell death by cell cycle analysis; gene expression by Affimetrix arrays and quantitative polymerase chain reaction; NF-κB activity by reporter assays; and protein levels by immunoblotting and immunofluorescence staining. We tested Tanespimycin/17-AAG and Fasudil treatment in groups of nine to 10 mice bearing ISOS-1 angiosarcoma. All statistical tests were two-sided. Results We discovered that DLC1 is a critical regulator of cell contact inhibition of proliferation in endothelial cells, promoting statistically significant (P < .001) cell death when cells are confluent (mean [SD] % viability: control DLC1 = 15.6 [19.3]; shDLC1 = 73.4 [13.1]). This prosurvival phenotype of DLC1-depleted confluent endothelial cells is attributable to a statistically significant and sustained increase of NF-κB activity (day 5, P = .001; day 8, P = .03) associated with increased tumor necrosis factor alpha-induced protein 3 (TNFAIP3/A20) signaling. Consistently, we found that DLC1 is statistically significantly reduced (P < .001 in 5 of 6) and TNFAIP3/A20 is statistically significantly increased (P < .001 in 2 of 3 and P = 0.02 in 1 of 3) in human angiosarcoma compared with normal adjacent endothelium. Treatment with the NF-κB inhibitor Tanespimycin/17-AAG statistically significantly reduced angiosarcoma tumor growth in mice (treatment tumor weight vs control, 0.50 [0.19] g vs 0.91 [0.21] g, P = .001 experiment 1; 0.66 [0.26] g vs 1.10 [0.31] g, P = .01 experiment 2). Conclusions These results identify DLC1 as a previously unrecognized regulator of endothelial cell contact inhibition of proliferation that is depleted in angiosarcoma and support NF-κB targeting for the treatment of angiosarcoma where DLC1 is lost.
Collapse
Affiliation(s)
- David Sánchez-Martín
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Atsushi Otsuka
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Taekyu Ha
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Dunrui Wang
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Xiaolan Qian
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Douglas R Lowy
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
17
|
Yan C, Huang WY, Boudreau J, Mayavannan A, Cheng Z, Wang J. IL-17R deletion predicts high-grade colorectal cancer and poor clinical outcomes. Int J Cancer 2019; 145:548-558. [PMID: 30628053 DOI: 10.1002/ijc.32122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 12/10/2018] [Accepted: 12/20/2018] [Indexed: 01/07/2023]
Abstract
The IL-17 receptor (IL-17R) has a perplexing role in cancer, which may be explained by its yin-yang signaling pathways. Recently, the critical role of IL-17R in maintaining basal levels of A20-a key negative regulator of NF-κB and JNK-c-Jun pathways has been demonstrated in cancer cell lines. Cross-cancer analyses of somatic copy number alterations in IL-17RA, IL-17RC and A20 genes reveal that IL-17RA-deletion is common in colorectal cancer (CRC) patients, representing 24, 26, 37 and 49% of stage I, II, III and IV of patients, respectively, and mutually exclusive with patients displaying microsatellite instability. Importantly, patients with IL-17R-deletion or concurrent deletions of A20 show significantly reduced overall survival. Analysis of multiple published microarray studies confirms that IL-17RA expression is significantly reduced in CRC samples compared to normal counterparts, and its level is closely associated with A20 expression. Analyses of RNAseq data indicate that tumors with IL-17R-deletion express strong molecular markers of tumor invasion, growth and metastasis. Notably, approximately 20 genes responsible for protein synthesis and mitochondrial metabolism are inversely correlated with both IL-17RA and A20. Immunohistochemistry staining in human colorectal tissue arrays further reveals that high-grade tumors have significantly reduced IL-17RA staining compared to low-grade tumors. Thus, collective evidence strongly supports a previously unrecognized CRC-promoting mechanism triggered by IL-17RA-deletion and highlights its utility as a prognostic marker in CRC.
Collapse
Affiliation(s)
- Chi Yan
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.,Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.,Canadian Center for Vaccinology, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Weei-Yuan Huang
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jeanette Boudreau
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.,Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Animamalar Mayavannan
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.,Canadian Center for Vaccinology, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Zhenyu Cheng
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.,Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.,Canadian Center for Vaccinology, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Jun Wang
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.,Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.,Canadian Center for Vaccinology, IWK Health Centre, Halifax, Nova Scotia, Canada
| |
Collapse
|
18
|
Rajamäki K, Keskitalo S, Seppänen M, Kuismin O, Vähäsalo P, Trotta L, Väänänen A, Glumoff V, Keskitalo P, Kaarteenaho R, Jartti A, Hautala N, Jackson P, Nordström DC, Saarela J, Hautala T, Eklund KK, Varjosalo M. Haploinsufficiency of A20 impairs protein-protein interactome and leads into caspase-8-dependent enhancement of NLRP3 inflammasome activation. RMD Open 2018. [PMID: 30402268 DOI: 10.1136/rmdopen-2018-000740)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVES TNFAIP3 encodes A20 that negatively regulates nuclear factor kappa light chain enhancer of activated B cells (NF-κB), the major transcription factor coordinating inflammatory gene expression. TNFAIP3 polymorphisms have been linked with a spectrum of inflammatory and autoimmune diseases and, recently, loss-of-function mutations in A20 were found to cause a novel inflammatory disease 'haploinsufficiency of A20' (HA20). Here we describe a family with HA20 caused by a novel TNFAIP3 loss-of-function mutation and elucidate the upstream molecular mechanisms linking HA20 to dysregulation of NF-κB and the related inflammasome pathway. METHODS NF-κB activation was studied in a mutation-expressing cell line using luciferase reporter assay. Physical and close-proximity protein-protein interactions of wild-type and TNFAIP3 p.(Lys91*) mutant A20 were analysed using mass spectrometry. NF-κB -dependent transcription, cytokine secretion and inflammasome activation were compared in immune cells of the HA20 patients and control subjects. RESULTS The protein-protein interactome of p.(Lys91*) mutant A20 was severely impaired, including interactions with proteins regulating NF-κB activation, DNA repair responses and the NLR family pyrin domain containing 3 (NLRP3) inflammasome. The p.(Lys91*) mutant A20 failed to suppress NF-κB signalling, which led to increased NF-κB -dependent proinflammatory cytokine transcription. Functional experiments in the HA20 patients' immune cells uncovered a novel caspase-8-dependent mechanism of NLRP3 inflammasome hyperresponsiveness that mediated the excessive secretion of interleukin-1β and interleukin-18. CONCLUSIONS The current findings significantly deepen our understanding of the molecular mechanisms underlying HA20 and other diseases associated with reduced A20 expression or function, paving the way for future therapeutic targeting of the pathway.
Collapse
Affiliation(s)
- Kristiina Rajamäki
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Salla Keskitalo
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Mikko Seppänen
- Immunodeficiency Unit, Inflammation Center and Rare Diseases Center, Children's Hospital, Helsinki University and Helsinki University Hospital, Helsinki, Finland
| | - Outi Kuismin
- Department of Clinical Genetics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Paula Vähäsalo
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Luca Trotta
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Antti Väänänen
- Department of Infection Control, Lapland Central Hospital, Rovaniemi, Finland
| | - Virpi Glumoff
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Paula Keskitalo
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Riitta Kaarteenaho
- Respiratory Diseases, Research Unit of Internal Medicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital, Oulu, Finland
| | - Airi Jartti
- Department of Radiology, Oulu University Hospital, Oulu, Finland
| | - Nina Hautala
- Department of Ophthalmology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Päivi Jackson
- Department of Ophthalmology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Dan C Nordström
- Department of Medicine and Rehabilitation, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Janna Saarela
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Timo Hautala
- Research Unit of Internal Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Kari K Eklund
- Department of Rheumatology, Inflammation Center, Helsinki University and Helsinki University Hospital, Helsinki, Finland.,Research Institute, Invalid Foundation, Helsinki, Finland.,Orton Orthopaedic Hospital, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.,Proteomics Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
19
|
Rajamäki K, Keskitalo S, Seppänen M, Kuismin O, Vähäsalo P, Trotta L, Väänänen A, Glumoff V, Keskitalo P, Kaarteenaho R, Jartti A, Hautala N, Jackson P, Nordström DC, Saarela J, Hautala T, Eklund KK, Varjosalo M. Haploinsufficiency of A20 impairs protein-protein interactome and leads into caspase-8-dependent enhancement of NLRP3 inflammasome activation. RMD Open 2018; 4:e000740. [PMID: 30402268 PMCID: PMC6203104 DOI: 10.1136/rmdopen-2018-000740] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/03/2018] [Accepted: 09/07/2018] [Indexed: 01/11/2023] Open
Abstract
Objectives TNFAIP3 encodes A20 that negatively regulates nuclear factor kappa light chain enhancer of activated B cells (NF-κB), the major transcription factor coordinating inflammatory gene expression. TNFAIP3 polymorphisms have been linked with a spectrum of inflammatory and autoimmune diseases and, recently, loss-of-function mutations in A20 were found to cause a novel inflammatory disease ‘haploinsufficiency of A20’ (HA20). Here we describe a family with HA20 caused by a novel TNFAIP3 loss-of-function mutation and elucidate the upstream molecular mechanisms linking HA20 to dysregulation of NF-κB and the related inflammasome pathway. Methods NF-κB activation was studied in a mutation-expressing cell line using luciferase reporter assay. Physical and close-proximity protein–protein interactions of wild-type and TNFAIP3 p.(Lys91*) mutant A20 were analysed using mass spectrometry. NF-κB -dependent transcription, cytokine secretion and inflammasome activation were compared in immune cells of the HA20 patients and control subjects. Results The protein–protein interactome of p.(Lys91*) mutant A20 was severely impaired, including interactions with proteins regulating NF-κB activation, DNA repair responses and the NLR family pyrin domain containing 3 (NLRP3) inflammasome. The p.(Lys91*) mutant A20 failed to suppress NF-κB signalling, which led to increased NF-κB -dependent proinflammatory cytokine transcription. Functional experiments in the HA20 patients’ immune cells uncovered a novel caspase-8-dependent mechanism of NLRP3 inflammasome hyperresponsiveness that mediated the excessive secretion of interleukin-1β and interleukin-18. Conclusions The current findings significantly deepen our understanding of the molecular mechanisms underlying HA20 and other diseases associated with reduced A20 expression or function, paving the way for future therapeutic targeting of the pathway.
Collapse
Affiliation(s)
- Kristiina Rajamäki
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Salla Keskitalo
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Mikko Seppänen
- Immunodeficiency Unit, Inflammation Center and Rare Diseases Center, Children's Hospital, Helsinki University and Helsinki University Hospital, Helsinki, Finland
| | - Outi Kuismin
- Department of Clinical Genetics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Paula Vähäsalo
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Luca Trotta
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Antti Väänänen
- Department of Infection Control, Lapland Central Hospital, Rovaniemi, Finland
| | - Virpi Glumoff
- Research Unit of Biomedicine, University of Oulu, Oulu, Finland
| | - Paula Keskitalo
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Riitta Kaarteenaho
- Respiratory Diseases, Research Unit of Internal Medicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital, Oulu, Finland
| | - Airi Jartti
- Department of Radiology, Oulu University Hospital, Oulu, Finland
| | - Nina Hautala
- Department of Ophthalmology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Päivi Jackson
- Department of Ophthalmology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Dan C Nordström
- Department of Medicine and Rehabilitation, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Janna Saarela
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Timo Hautala
- Research Unit of Internal Medicine, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Kari K Eklund
- Department of Rheumatology, Inflammation Center, Helsinki University and Helsinki University Hospital, Helsinki, Finland.,Research Institute, Invalid Foundation, Helsinki, Finland.,Orton Orthopaedic Hospital, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland.,Proteomics Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
20
|
MyD88 signaling causes autoimmune sialadenitis through formation of high endothelial venules and upregulation of LTβ receptor-mediated signaling. Sci Rep 2018; 8:14272. [PMID: 30250175 PMCID: PMC6155371 DOI: 10.1038/s41598-018-32690-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 09/13/2018] [Indexed: 01/17/2023] Open
Abstract
Autoimmune sialadenitis (AS), chronic inflammation of the salivary glands (SGs) with focal lymphocyte infiltration, appears in autoimmune diseases such as Sjӧgren’s syndrome. The pathological role of MyD88-dependent innate immune signaling in autoimmune diseases including AS has been studied using mouse models, such as NOD mice. Although AS development in NOD mice was reported to be suppressed by Myd88 deficiency, its specific role remains unclear. Here, we determined the potent suppressive effects of Myd88 deficiency on AS development in lupus-prone B6/lpr mice, which have lymphoproliferation abnormalities, and also in NOD mice, which have no lymphoproliferation abnormalities. This indicates that MyD88 signaling triggers AS through both lymphoproliferation-dependent and -independent mechanisms. To address the MyD88-dependent lymphoproliferation-independent AS manifestation, SGs from C57BL/6 mice were analyzed. Remarkable upregulation of Glycam1 and high endothelial venule (HEV)-associated changes were unexpectedly found in Myd88+/+ mice, compared with Myd88−/− mice. MyD88-dependent HEV-associated changes were also observed in NOD mice. Additionally, Lta, Ltb, and Ltbr in SGs of NOD mice were lowered by Myd88 deficiency. Interestingly, LTβR-induced HEV-associated gene expression in cultured cells was impaired by Myd88 deficiency. Our findings highlight novel roles for MyD88 in AS development, which imply the existence of MyD88-dependent HEV formation in ectopic lymphoid neogenesis.
Collapse
|
21
|
Struzik J, Szulc-Dąbrowska L. Manipulation of Non-canonical NF-κB Signaling by Non-oncogenic Viruses. Arch Immunol Ther Exp (Warsz) 2018; 67:41-48. [PMID: 30196473 PMCID: PMC6433803 DOI: 10.1007/s00005-018-0522-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023]
Abstract
Nuclear factor (NF)-κB is a major regulator of antiviral response. Viral pathogens exploit NF-κB activation pathways to avoid cellular mechanisms that eliminate the infection. Canonical (classical) NF-κB signaling, which regulates innate immune response, cell survival and inflammation, is often manipulated by viral pathogens that can counteract antiviral response. Oncogenic viruses can modulate not only canonical, but also non-canonical (alternative) NF-κB activation pathways. The non-canonical NF-κB signaling is responsible for adaptive immunity and plays a role in lymphoid organogenesis, B cell development, as well as bone metabolism. Thus, non-canonical NF-κB activation has been linked to lymphoid malignancies. However, some data strongly suggest that the non-canonical NF-κB activation pathway may also function in innate immunity and is modulated by certain non-oncogenic viruses. Collectively, these findings show the importance of studying the impact of different groups of viral pathogens on alternative NF-κB activation. This mini-review focuses on the influence of non-oncogenic viruses on the components of non-canonical NF-κB signaling.
Collapse
Affiliation(s)
- Justyna Struzik
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8, 02-786, Warsaw, Poland.
| | - Lidia Szulc-Dąbrowska
- Division of Immunology, Department of Preclinical Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8, 02-786, Warsaw, Poland
| |
Collapse
|
22
|
Rothschild DE, McDaniel DK, Ringel-Scaia VM, Allen IC. Modulating inflammation through the negative regulation of NF-κB signaling. J Leukoc Biol 2018; 103:10.1002/JLB.3MIR0817-346RRR. [PMID: 29389019 PMCID: PMC6135699 DOI: 10.1002/jlb.3mir0817-346rrr] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/16/2022] Open
Abstract
Immune system activation is essential to thwart the invasion of pathogens and respond appropriately to tissue damage. However, uncontrolled inflammation can result in extensive collateral damage underlying a diverse range of auto-inflammatory, hyper-inflammatory, and neoplastic diseases. The NF-κB signaling pathway lies at the heart of the immune system and functions as a master regulator of gene transcription. Thus, this signaling cascade is heavily targeted by mechanisms designed to attenuate overzealous inflammation and promote resolution. Mechanisms associated with the negative regulation of NF-κB signaling are currently under intense investigation and have yet to be fully elucidated. Here, we provide an overview of mechanisms that negatively regulate NF-κB signaling through either attenuation of signal transduction, inhibition of posttranscriptional signaling, or interference with posttranslational modifications of key pathway components. While the regulators discussed for each group are far from comprehensive, they exemplify common mechanistic approaches that inhibit this critical biochemical signaling cascade. Despite their diversity, a commonality among these regulators is their selection of specific targets at key inflection points in the pathway, such as TNF-receptor-associated factor family members or essential kinases. A better understanding of these negative regulatory mechanisms will be essential to gain greater insight related to the maintenance of immune system homeostasis and inflammation resolution. These processes are vital elements of disease pathology and have important implications for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Daniel E. Rothschild
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg VA 24061
| | - Dylan K. McDaniel
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg VA 24061
| | - Veronica M. Ringel-Scaia
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg VA 24061
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016
| |
Collapse
|
23
|
Clancy-Thompson E, Ali L, Bruck PT, Exley MA, Blumberg RS, Dranoff G, Dougan M, Dougan SK. IAP Antagonists Enhance Cytokine Production from Mouse and Human iNKT Cells. Cancer Immunol Res 2018; 6:25-35. [PMID: 29187357 PMCID: PMC5754232 DOI: 10.1158/2326-6066.cir-17-0490] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/18/2017] [Accepted: 11/22/2017] [Indexed: 01/05/2023]
Abstract
Inhibitor of apoptosis protein (IAP) antagonists are in clinical trials for a variety of cancers, and mouse models show synergism between IAP antagonists and anti-PD-1 immunotherapy. Although IAP antagonists affect the intrinsic signaling of tumor cells, their most pronounced effects are on immune cells and the generation of antitumor immunity. Here, we examined the effects of IAP antagonism on T-cell development using mouse fetal thymic organ culture and observed a selective loss of iNKT cells, an effector cell type of potential importance for cancer immunotherapy. Thymic iNKT-cell development probably failed due to increased strength of TCR signal leading to negative selection, given that mature iNKT cells treated with IAP antagonists were not depleted, but had enhanced cytokine production in both mouse and human ex vivo cultures. Consistent with this, mature mouse primary iNKT cells and iNKT hybridomas increased production of effector cytokines in the presence of IAP antagonists. In vivo administration of IAP antagonists and α-GalCer resulted in increased IFNγ and IL-2 production from iNKT cells and decreased tumor burden in a mouse model of melanoma lung metastasis. Human iNKT cells also proliferated and increased IFNγ production dramatically in the presence of IAP antagonists, demonstrating the utility of these compounds in adoptive therapy of iNKT cells. Cancer Immunol Res; 6(1); 25-35. ©2017 AACR.
Collapse
Affiliation(s)
- Eleanor Clancy-Thompson
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lestat Ali
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patrick T Bruck
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark A Exley
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Richard S Blumberg
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Glenn Dranoff
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michael Dougan
- Harvard Medical School, Boston, Massachusetts.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
24
|
Osako M, Itsumi M, Yamaguchi H, Takeuchi H, Yamaoka S. A20 restores phorbol ester-induced differentiation of THP-1 cells in the absence of nuclear factor-κB activation. J Cell Biochem 2017; 119:1475-1487. [PMID: 28771803 DOI: 10.1002/jcb.26308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 08/02/2017] [Indexed: 11/06/2022]
Abstract
A20, also referred to as tumor necrosis factor alpha (TNFα)-induced protein 3 (TNFAIP3), is an ubiquitin-editing enzyme whose expression is enhanced by NF-κB activation, and plays an important role in silencing NF-κB activity. Another well-known role for A20 is to protect cells from TNFα-induced apoptosis. Depletion of NF-κB in differentiating U937 monocytic leukemia cells is known to cause apoptotic cell death; however, much remains to be explored about the molecules that are expressed in an NF-κB-dependent manner and which support monocyte-macrophage differentiation. Using the monocytic cell line THP-1, and peripheral blood monocytes, we show here a sustained increase in A20 expression during monocyte-macrophage differentiation, which coincided with high NF-κB-dependent transcriptional activity. Depletion of NF-κB by stable expression of a super-repressor form of IκBα in THP-1 cells caused remarkable cell death during phorbol 12-myristate 13-acetate (PMA)-induced differentiation. A20 expression in these cells did not alter this NF-κB suppression, but was sufficient to protect the cells and restore the cell surface expression of a differentiation marker (CD11b) and phagocytic activity. Mutational analyses revealed that this A20 activity requires the carboxy-terminal zinc-finger domain, but not its deubiquitinase activity. Based on these findings, we conclude that A20, when ectopically expressed, can support both survival and differentiation of THP-1 cells in the absence of sustained NF-κB activity.
Collapse
Affiliation(s)
- Miho Osako
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Momoe Itsumi
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Haruka Yamaguchi
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hiroaki Takeuchi
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Shoji Yamaoka
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
25
|
Song ZX, Jiang WD, Liu Y, Wu P, Jiang J, Zhou XQ, Kuang SY, Tang L, Tang WN, Zhang YA, Feng L. Dietary zinc deficiency reduced growth performance, intestinal immune and physical barrier functions related to NF-κB, TOR, Nrf2, JNK and MLCK signaling pathway of young grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2017; 66:497-523. [PMID: 28549941 DOI: 10.1016/j.fsi.2017.05.048] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/11/2017] [Accepted: 05/19/2017] [Indexed: 06/07/2023]
Abstract
Our study investigated the effects of dietary zinc (Zn) deficiency on growth performance, intestinal immune and physical barrier functions of young grass carp (Ctenopharyngodon idella). A total of 630 grass carp (244.14 ± 0.40 g) were fed graded levels of zinc lactate (10.71, 30.21, 49.84, 72.31, 92.56, 110.78 mg Zn/kg diet) and one zinc sulfate group (56.9 mg Zn/kg diet) for 60 days. At the end of the feeding trial, fish were challenged with Aeromonas hydrophila for 14 days. These results indicated that compared with optimal dietary Zn level, dietary Zn deficiency (10.71 mg/kg diet) decreased the production of antibacterial compounds, up-regulated pro-inflammatory cytokines related to nuclear factor kappa B (NF-κB) and down-regulated anti-inflammatory cytokines related to target of rapamycin (TOR) in three intestinal segments of young grass carp (P < 0.05), suggesting that dietary Zn deficiency could impair intestinal immune barrier of fish; decreased the activities and mRNA levels of antioxidant enzymes related to NF-E2-related factor 2 (Nrf2), up-regulated the mRNA levels of caspase-3, -7, -8, -9 related to p38 mitogen activated protein (p38 MAPK) and c-Jun N-terminal protein kinase (JNK), down-regulated the mRNA levels of tight junction complexes (TJs) related to myosin light chain kinase (MLCK) in three intestinal segments of young grass carp (P < 0.05), demonstrating that dietary Zn deficiency could injury intestinal physical barrier of fish. Besides, the Zn requirements (zinc lactate as Zn source) based on percent weight gain (PWG), against enteritis morbidity, acid phosphatase (ACP) activity in the proximal intestine (PI) and malondialdehyde (MDA) content in the PI of young grass carp was estimated to be 61.2, 61.4, 69.2 and 69.5 mg/kg diet, respectively. Finally, based on specific growth rate (SGR), feed efficiency (FE) and against enteritis morbidity of young grass carp, the efficacy of zinc lactate relative to zinc sulfate were 132.59%, 135.27% and 154.04%, respectively.
Collapse
Affiliation(s)
- Zheng-Xing Song
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Wu-Neng Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Sichuan, Chengdu 611130, China.
| |
Collapse
|
26
|
Uematsu M, Nishimura T, Sakamaki Y, Yamamoto H, Mizushima N. Accumulation of undegraded autophagosomes by expression of dominant-negative STX17 (syntaxin 17) mutants. Autophagy 2017; 13:1452-1464. [PMID: 28598244 DOI: 10.1080/15548627.2017.1327940] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Macroautophagy/autophagy, which is one of the main degradation systems in the cell, is mediated by a specialized organelle, the autophagosome. Purification of autophagosomes before fusion with lysosomes is important for both mechanistic and physiological studies of the autophagosome. Here, we report a simple method to accumulate undigested autophagosomes. Overexpression of the autophagosomal Qa-SNARE STX17 (syntaxin 17) lacking the N-terminal domain (NTD) or N-terminally tagged GFP-STX17 causes accumulation of autophagosomes. A HeLa cell line, which expresses GFP-STX17ΔNTD or full-length GFP-STX17 under the control of the tetracycline-responsive promoter, accumulates a large number of undigested autophagosomes devoid of lysosomal markers or early autophagy factors upon treatment with doxycycline. Using this inducible cell line, nascent autophagosomes can be easily purified by OptiPrep density-gradient centrifugation and immunoprecipitation. This novel method should be useful for further characterization of nascent autophagosomes.
Collapse
Affiliation(s)
- Masaaki Uematsu
- a Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine , The University of Tokyo , Tokyo , Japan
| | - Taki Nishimura
- a Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine , The University of Tokyo , Tokyo , Japan
| | - Yuriko Sakamaki
- b Research Center for Medical and Dental Sciences , Tokyo Medical and Dental University , Tokyo , Japan
| | - Hayashi Yamamoto
- a Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine , The University of Tokyo , Tokyo , Japan
| | - Noboru Mizushima
- a Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine , The University of Tokyo , Tokyo , Japan
| |
Collapse
|
27
|
Nishimura T, Tamura N, Kono N, Shimanaka Y, Arai H, Yamamoto H, Mizushima N. Autophagosome formation is initiated at phosphatidylinositol synthase-enriched ER subdomains. EMBO J 2017; 36:1719-1735. [PMID: 28495679 DOI: 10.15252/embj.201695189] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 03/28/2017] [Accepted: 04/11/2017] [Indexed: 11/09/2022] Open
Abstract
The autophagosome, a double-membrane structure mediating degradation of cytoplasmic materials by macroautophagy, is formed in close proximity to the endoplasmic reticulum (ER). However, how the ER membrane is involved in autophagy initiation and to which membrane structures the autophagy-initiation complex is localized have not been fully characterized. Here, we were able to biochemically analyze autophagic intermediate membranes and show that the autophagy-initiation complex containing ULK and FIP200 first associates with the ER membrane. To further characterize the ER subdomain, we screened phospholipid biosynthetic enzymes and found that the autophagy-initiation complex localizes to phosphatidylinositol synthase (PIS)-enriched ER subdomains. Then, the initiation complex translocates to the ATG9A-positive autophagosome precursors in a PI3P-dependent manner. Depletion of phosphatidylinositol (PI) by targeting bacterial PI-specific phospholipase C to the PIS domain impairs recruitment of downstream autophagy factors and autophagosome formation. These findings suggest that the autophagy-initiation complex, the PIS-enriched ER subdomain, and ATG9A vesicles together initiate autophagosome formation.
Collapse
Affiliation(s)
- Taki Nishimura
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Norito Tamura
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Nozomu Kono
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuta Shimanaka
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Arai
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Hayashi Yamamoto
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
Zeng P, Ma J, Yang R, Liu YC. Immune Regulation by Ubiquitin Tagging as Checkpoint Code. Curr Top Microbiol Immunol 2017; 410:215-248. [PMID: 28929193 DOI: 10.1007/82_2017_64] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The immune system is equipped with effective machinery to mobilize its activation to defend invading microorganisms, and at the same time, to refrain from attacking its own tissues to maintain immune tolerance. The balance of activation and tolerance is tightly controlled by diverse mechanisms, since breakdown of tolerance could result in disastrous consequences such as the development of autoimmune diseases. One of the mechanisms is by the means of protein ubiquitination, which involves the process of tagging a small peptide ubiquitin to protein substrates. E3 ubiquitin ligases are responsible for catalyzing the final step of ubiquitin-substrate conjugation by specifically recognizing substrates to determine their fates of degradation or functional modification. The ubiquitination process is reversible, which is carried out by deubiquitinating enzymes to release the ubiquitin molecule from the conjugated substrates. Protein ubiquitination and deubiquitination serve as checkpoint codes in many key steps of lymphocyte regulation including the development, activation, differentiation, and tolerance induction. In this chapter, we will discuss a few E3 ligases and deubiquitinating enzymes that are important in controlling immune responses, with emphasis on their roles in T cells.
Collapse
Affiliation(s)
- Peng Zeng
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Jieyu Ma
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Runqing Yang
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yun-Cai Liu
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China. .,Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037, USA.
| |
Collapse
|
29
|
Yamaguchi N, Shibazaki M, Yamada C, Anzai E, Morii M, Nakayama Y, Kuga T, Hashimoto Y, Tomonaga T, Yamaguchi N. Tyrosine Phosphorylation of the Pioneer Transcription Factor FoxA1 Promotes Activation of Estrogen Signaling. J Cell Biochem 2016; 118:1453-1461. [DOI: 10.1002/jcb.25804] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 11/21/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Noritaka Yamaguchi
- Laboratory of Molecular Cell BiologyGraduate School of Pharmaceutical SciencesChiba UniversityChiba260‐8675Japan
| | - Misato Shibazaki
- Laboratory of Molecular Cell BiologyGraduate School of Pharmaceutical SciencesChiba UniversityChiba260‐8675Japan
| | - Chiaki Yamada
- Laboratory of Molecular Cell BiologyGraduate School of Pharmaceutical SciencesChiba UniversityChiba260‐8675Japan
| | - Erina Anzai
- Laboratory of Molecular Cell BiologyGraduate School of Pharmaceutical SciencesChiba UniversityChiba260‐8675Japan
| | - Mariko Morii
- Laboratory of Molecular Cell BiologyGraduate School of Pharmaceutical SciencesChiba UniversityChiba260‐8675Japan
| | - Yuji Nakayama
- Department of Biochemistry and Molecular BiologyKyoto Pharmaceutical UniversityKyoto607‐8414Japan
| | - Takahisa Kuga
- Department of Biochemistry and Molecular BiologyKyoto Pharmaceutical UniversityKyoto607‐8414Japan
| | - Yuuki Hashimoto
- Laboratory of Proteome ResearchNational Institutes of Biomedical InnovationHealth and NutritionIbarakiOsaka567‐0085Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome ResearchNational Institutes of Biomedical InnovationHealth and NutritionIbarakiOsaka567‐0085Japan
| | - Naoto Yamaguchi
- Laboratory of Molecular Cell BiologyGraduate School of Pharmaceutical SciencesChiba UniversityChiba260‐8675Japan
| |
Collapse
|
30
|
M1 and M2 Functional Imprinting of Primary Microglia: Role of P2X7 Activation and miR-125b. Mediators Inflamm 2016; 2016:2989548. [PMID: 28090150 PMCID: PMC5206439 DOI: 10.1155/2016/2989548] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/24/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a most frequently occurring and severe form of motor neuron disease, causing death within 3-5 years from diagnosis and with a worldwide incidence of about 2 per 100,000 person-years. Mutations in over twenty genes associated with familial forms of ALS have provided insights into the mechanisms leading to motor neuron death. Moreover, mutations in two RNA binding proteins, TAR DNA binding protein 43 and fused in sarcoma, have raised the intriguing possibility that perturbations of RNA metabolism, including that of the small endogenous RNA molecules that repress target genes at the posttranscriptional level, that is, microRNAs, may contribute to disease pathogenesis. At present, the mechanisms by which microglia actively participate to both toxic and neuroprotective actions in ALS constitute an important matter of research. Among the pathways involved in ALS-altered microglia responses, in previous works we have uncovered the hyperactivation of P2X7 receptor by extracellular ATP and the overexpression of miR-125b, both leading to uncontrolled toxic M1 reactions. In order to shed further light on the complexity of these processes, in this short review we will describe the M1/M2 functional imprinting of primary microglia and a role played by P2X7 and miR-125b in ALS microglia activation.
Collapse
|
31
|
Jin WL, Mao XY, Qiu GZ. Targeting Deubiquitinating Enzymes in Glioblastoma Multiforme: Expectations and Challenges. Med Res Rev 2016; 37:627-661. [PMID: 27775833 DOI: 10.1002/med.21421] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 09/06/2016] [Accepted: 09/25/2016] [Indexed: 12/16/2022]
Abstract
Glioblastoma (GBM) is regarded as the most common primary intracranial neoplasm. Despite standard treatment with tumor resection and radiochemotherapy, the outcome remains gloomy. It is evident that a combination of oncogenic gain of function and tumor-suppressive loss of function has been attributed to glioma initiation and progression. The ubiquitin-proteasome system is a well-orchestrated system that controls the fate of most proteins by striking a dynamic balance between ubiquitination and deubiquitination of substrates, having a profound influence on the modulation of oncoproteins, tumor suppressors, and cellular signaling pathways. In recent years, deubiquitinating enzymes (DUBs) have emerged as potential anti-cancer targets due to their targeting several key proteins involved in the regulation of tumorigenesis, apoptosis, senescence, and autophagy. This review attempts to summarize recent studies of GBM-associated DUBs, their roles in various cellular processes, and discuss the relation between DUBs deregulation and gliomagenesis, especially how DUBs regulate glioma stem cells pluripotency, microenvironment, and resistance of radiation and chemotherapy through core stem-cell transcriptional factors. We also review recent achievements and progress in the development of potent and selective reversible inhibitors of DUBs, and attempted to find a potential GBM treatment by DUBs intervention.
Collapse
Affiliation(s)
- Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China.,National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, P. R. China
| | - Guan-Zhong Qiu
- Department of Neurosurgery, General Hospital of Jinan Military Command, Jinan, 250031, P. R. China
| |
Collapse
|
32
|
TRAF2 multitasking in TNF receptor-induced signaling to NF-κB, MAP kinases and cell death. Biochem Pharmacol 2016; 116:1-10. [DOI: 10.1016/j.bcp.2016.03.009] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 03/14/2016] [Indexed: 12/15/2022]
|
33
|
McDaniel DK, Eden K, Ringel VM, Allen IC. Emerging Roles for Noncanonical NF-κB Signaling in the Modulation of Inflammatory Bowel Disease Pathobiology. Inflamm Bowel Dis 2016; 22:2265-79. [PMID: 27508514 PMCID: PMC4992436 DOI: 10.1097/mib.0000000000000858] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Crohn's disease and ulcerative colitis are common and debilitating manifestations of inflammatory bowel disease (IBD). IBD is characterized by a radical imbalance in the activation of proinflammatory and anti-inflammatory signaling pathways in the gut. These pathways are controlled by NF-κB, which is a master regulator of gene transcription. In IBD patients, NF-κB signaling is often dysregulated resulting in overzealous inflammation. NF-κB activation occurs through 2 distinct pathways, defined as either canonical or noncanonical. Canonical NF-κB pathway activation is well studied in IBD and is associated with the rapid, acute production of diverse proinflammatory mediators, such as COX-2, IL-1β, and IL-6. In contrast to the canonical pathway, the noncanonical or "alternative" NF-κB signaling cascade is tightly regulated and is responsible for the production of highly specific chemokines that tend to be associated with less acute, chronic inflammation. There is a relative paucity of literature regarding all aspects of noncanonical NF-ĸB signaling. However, it is clear that this alternative signaling pathway plays a considerable role in maintaining immune system homeostasis and likely contributes significantly to the chronic inflammation underlying IBD. Noncanonical NF-κB signaling may represent a promising new direction in the search for therapeutic targets and biomarkers associated with IBD. However, significant mechanistic insight is still required to translate the current basic science findings into effective therapeutic strategies.
Collapse
Affiliation(s)
- Dylan K. McDaniel
- Department of Biomedical Sciences and Pathobiology, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061
| | - Kristin Eden
- Department of Biomedical Sciences and Pathobiology, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061
| | - Veronica M. Ringel
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061
| |
Collapse
|
34
|
Abstract
High levels of miR-23a expression in human primary macrophages suggested that miR-23a might have an important role in innate immune cells. Therefore, we investigated whether miR-23a regulates the secretion of cytokines by immune cells. Herein, we demonstrate that the expression of miR-23a was reduced in toll-like receptor (TLR)-stimulated macrophages. By targeting A20, miR-23a could affect NF-κB activity and the expression of downstream NF-κB-target genes that encode proinflammatory mediators, such as IL-6 and TNF-α. In summary, our study describes a novel role for miR-23a in the regulation of inflammatory cytokine production in macrophages, which could yield new insights into the regulatory role of miRNAs in the inflammatory response.
Collapse
|
35
|
Liu X, Liu Y, Xu M, Li J, Teng X, Cheng H, Xia Y. Zinc finger protein A20 is involved in the antipsoriatic effect of calcipotriol. Br J Dermatol 2016; 175:314-24. [PMID: 26875609 DOI: 10.1111/bjd.14481] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2016] [Indexed: 02/05/2023]
Affiliation(s)
- X. Liu
- Department of Dermatology; The Third Affiliated Hospital of Soochow University; Changzhou China
| | - Y. Liu
- Department of Dermatology; The Second Affiliated Hospital; School of Medicine; Xi'an Jiaotong University; Xi'an 710004 China
| | - M. Xu
- Department of Dermatology; The Second Affiliated Hospital; School of Medicine; Xi'an Jiaotong University; Xi'an 710004 China
| | - J. Li
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy; West China Hospital; Sichuan University; Chengdu China
| | - X. Teng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy; West China Hospital; Sichuan University; Chengdu China
| | - H. Cheng
- Department of Medicine; The Second Affiliated Hospital; School of Medicine; Xi'an Jiaotong University; Xi'an 710004 China
| | - Y. Xia
- Department of Dermatology; The Second Affiliated Hospital; School of Medicine; Xi'an Jiaotong University; Xi'an 710004 China
| |
Collapse
|
36
|
Saitoh Y, Hamano A, Mochida K, Kakeya A, Uno M, Tsuruyama E, Ichikawa H, Tokunaga F, Utsunomiya A, Watanabe T, Yamaoka S. A20 targets caspase-8 and FADD to protect HTLV-I-infected cells. Leukemia 2015; 30:716-27. [PMID: 26437781 DOI: 10.1038/leu.2015.267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 08/31/2015] [Accepted: 09/18/2015] [Indexed: 12/15/2022]
Abstract
Adult T-cell leukemia (ATL) arises from a human T-cell leukemia virus type I (HTLV-I)-infected cell and has few therapeutic options. Here, we have uncovered a previously unrecognized role for a ubiquitin-editing enzyme A20 in the survival of HTLV-I-infected cells. Unlike in lymphomas of the B-cell lineage, A20 is abundantly expressed in primary ATL cells without notable mutations. Depletion of A20 in HTLV-I-infected cells resulted in caspase activation, cell death induction and impaired tumorigenicity in mouse xenograft models. Mechanistically, A20 stably interacts with caspase-8 and Fas-associated via death domain (FADD) in HTLV-I-infected cells. Mutational studies revealed that A20 supports the growth of HTLV-I-infected cells independent of its catalytic functions and that the zinc-finger domains are required for the interaction with and regulation of caspases. These results indicate a pivotal role for A20 in the survival of HTLV-I-infected cells and implicate A20 as a potential therapeutic target in ATL.
Collapse
Affiliation(s)
- Y Saitoh
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - A Hamano
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - K Mochida
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - A Kakeya
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - M Uno
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Comprehensive Reproductive Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - E Tsuruyama
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - H Ichikawa
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - F Tokunaga
- Laboratory of Molecular Cell Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - A Utsunomiya
- Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan
| | - T Watanabe
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - S Yamaoka
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
37
|
Verhelst K, Gardam S, Borghi A, Kreike M, Carpentier I, Beyaert R. XEDAR activates the non-canonical NF-κB pathway. Biochem Biophys Res Commun 2015; 465:275-80. [PMID: 26260321 DOI: 10.1016/j.bbrc.2015.08.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/04/2015] [Indexed: 01/12/2023]
Abstract
Members of the tumor necrosis factor receptor (TNFR) superfamily are involved in a number of physiological and pathological responses by activating a wide variety of intracellular signaling pathways. The X-linked ectodermal dysplasia receptor (XEDAR; also known as EDA2R or TNFRSF27) is a member of the TNFR superfamily that is highly expressed in ectodermal derivatives during embryonic development and binds to ectodysplasin-A2 (EDA-A2), a member of the TNF family that is encoded by the anhidrotic ectodermal dysplasia (EDA) gene. Although XEDAR was first described in the year 2000, its function and molecular mechanism of action is still largely unclear. XEDAR has been reported to activate canonical nuclear factor κB (NF-κB) signaling and mitogen-activated protein (MAP) kinases. Here we report that XEDAR is also able to trigger the non-canonical NF-κB pathway, characterized by the processing of p100 (NF-κB2) into p52, followed by nuclear translocation of p52 and RelB. We provide evidence that XEDAR-induced p100 processing relies on the binding of XEDAR to TRAF3 and TRAF6, and requires the kinase activity of NIK and IKKα. We also show that XEDAR stimulation results in NIK accumulation and that p100 processing is negatively regulated by TRAF3, cIAP1 and A20.
Collapse
Affiliation(s)
- Kelly Verhelst
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Sandra Gardam
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Alice Borghi
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Marja Kreike
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Isabelle Carpentier
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Rudi Beyaert
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
38
|
Lu H, Lei X, Zhang Q. Moderate activation of IKK2-NF-kB in unstressed adult mouse liver induces cytoprotective genes and lipogenesis without apparent signs of inflammation or fibrosis. BMC Gastroenterol 2015. [PMID: 26219821 PMCID: PMC4518658 DOI: 10.1186/s12876-015-0325-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background The NF-kB signaling, regulated by IKK1-p52/RelB and IKK2-p65, is activated by various stresses to protect or damage the liver, in context-specific manners. Two previous studies of liver-specific expression of constitutive active IKK2 (IKK2ca) showed that strong activation of IKK2-NF-kB in mouse livers caused inflammation, insulin resistance, and/or fibrosis. The purpose of this study was to understand how moderate activation of IKK2-NF-kB in adult mouse livers alters hepatic gene expression and pathophysiology. Method We generated mice with adult hepatocyte-specific activation of Ikk2 (Liv-Ikk2ca) using Alb-cre mice and Ikk2ca Rosa26 knockin mice in which a moderate expression of Ikk2ca transgene was driven by the endogenous Rosa26 promoter. Results Surprisingly, compared to wild-type mice, adult male Liv-Ikk2ca mice had higher hepatic mRNA expression of Ikk2 and classical NF-kB targets (e.g. Lcn2 and A20), as well as IKK1, NIK, and RelB, but no changes in markers of inflammation or fibrosis. Blood levels of IL-6 and MCP-1 remained unchanged, and histology analysis showed a lack of injury or infiltration of inflammatory cells in livers of Liv-Ikk2ca mice. Moreover, Liv-Ikk2ca mice had lower mRNA expression of prooxidative enzymes Cyp2e1 and Cyp4a14, higher expression of antioxidative enzymes Sod2, Gpx1, and Nqo1, without changes in key enzymes for fatty acid oxidation, glucose utilization, or gluconeogenesis. In parallel, Liv-Ikk2ca mice and wild-type mice had similar levels of hepatic reduced glutathione, endogenous reactive oxygen species, and lipid peroxidation. Additionally, Liv-Ikk2ca mice had higher Cyp3a11 without down-regulation of most drug processing genes. Regarding nuclear proteins of NF-kB subunits, Liv-Ikk2ca mice had moderately higher p65 and p50 but much higher RelB. Results of ChIP-qPCR showed that the binding of p50 to multiple NF-kB-target genes was markedly increased in Liv-Ikk2ca mice. Additionally, Liv-Ikk2ca mice had moderate increase in triglycerides in liver, which was associated with higher lipogenic factors Pparγ, Lxr, Fasn, Scd1, and CD36. Conclusion In summary, moderate activation of IKK2-NF-kB in unstressed adult mouse hepatocytes produces a cytoprotective gene expression profile and induces lipogenesis without apparent signs of inflammation or fibrosis, likely due to strong activation of the anti-inflammatory IKK1-RelB alternative NF-kB pathway as well as the Lxr. Electronic supplementary material The online version of this article (doi:10.1186/s12876-015-0325-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, 750 E Adams ST, Syracuse, NY, 13210, USA.
| | - Xiaohong Lei
- Department of Pharmacology, SUNY Upstate Medical University, 750 E Adams ST, Syracuse, NY, 13210, USA.
| | - Qinghao Zhang
- Department of Pharmacology, SUNY Upstate Medical University, 750 E Adams ST, Syracuse, NY, 13210, USA.
| |
Collapse
|
39
|
c-Abl-mediated tyrosine phosphorylation of JunB is required for Adriamycin-induced expression of p21. Biochem J 2015. [PMID: 26217035 DOI: 10.1042/bj20150372] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The non-receptor-type tyrosine kinase c-Abl functions as a cytoplasmic signal transducer upon activation of cell-surface receptors. c-Abl is also involved in DDR (DNA-damage response), which is initiated in the nucleus, whereas its molecular functions in DDR are not fully understood. In the present study, we found that c-Abl phosphorylates JunB, a member of the AP-1 (activator protein 1) transcription factor family. Because JunB was suggested to be involved in DDR, we analysed the role of c-Abl-mediated phosphorylation of JunB in DDR. We first analysed phosphorylation sites of JunB and found that c-Abl majorly phosphorylates JunB at Tyr(173), Tyr(182) and Tyr(188). Because c-Abl promotes expression of the cyclin-dependent kinase inhibitor p21 upon stimulation with the DNA-damaging agent Adriamycin (doxorubicin), we analysed the involvement of JunB in Adriamycin-induced p21 expression. We found that JunB suppresses p21 induction through inhibition of its promoter activity. The phosphomimetic JunB, which was generated by glutamic acid substitutions at the phosphorylation sites, failed to repress p21 induction. Recruitment of JunB to the p21 promoter was promoted by Adriamycin stimulation and was further enhanced by co-treatment with the c-Abl inhibitor imatinib. The phosphomimetic glutamic acid substitutions in JunB or Adriamycin treatment impaired the JunB-c-Fos transcription factor complex formation. Taken together, these results suggest that, although JunB represses p21 promoter activity, c-Abl phosphorylates JunB and conversely inhibits its suppressive role on p21 promoter activity upon Adriamycin stimulation. Therefore JunB is likely to be a key target of c-Abl in expression of p21 in Adriamycin-induced DDR.
Collapse
|
40
|
Shinzawa M, Konno H, Qin J, Akiyama N, Miyauchi M, Ohashi H, Miyamoto-Sato E, Yanagawa H, Akiyama T, Inoue JI. Catalytic subunits of the phosphatase calcineurin interact with NF-κB-inducing kinase (NIK) and attenuate NIK-dependent gene expression. Sci Rep 2015; 5:10758. [PMID: 26029823 PMCID: PMC5377069 DOI: 10.1038/srep10758] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 04/28/2015] [Indexed: 01/09/2023] Open
Abstract
Nuclear factor (NF)-κB-inducing kinase (NIK) is a serine/threonine kinase that activates NF-κB pathways, thereby regulating a wide variety of immune systems. Aberrant NIK activation causes tumor malignancy, suggesting a requirement for precise regulation of NIK activity. To explore novel interacting proteins of NIK, we performed in vitro virus screening and identified the catalytic subunit Aα isoform of serine/threonine phosphatase calcineurin (CnAα) as a novel NIK-interacting protein. The interaction of NIK with CnAα in living cells was confirmed by co-immunoprecipitation. Calcineurin catalytic subunit Aβ isoform (CnAβ) also bound to NIK. Experiments using domain deletion mutants suggested that CnAα and CnAβ interact with both the kinase domain and C-terminal region of NIK. Moreover, the phosphatase domain of CnAα is responsible for the interaction with NIK. Intriguingly, we found that TRAF3, a critical regulator of NIK activity, also binds to CnAα and CnAβ. Depletion of CnAα and CnAβ significantly enhanced lymphotoxin-β receptor (LtβR)-mediated expression of the NIK-dependent gene Spi-B and activation of RelA and RelB, suggesting that CnAα and CnAβ attenuate NF-κB activation mediated by LtβR-NIK signaling. Overall, these findings suggest a possible role of CnAα and CnAβ in modifying NIK functions.
Collapse
Affiliation(s)
- Miho Shinzawa
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Hiroyasu Konno
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Junwen Qin
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Department of Developmental and Regenerative Biology, Key Laboratory for Regenerative Medicine, Ministry of Education and International Base of Collaboration for Science and Technology, Ministry of Science and Technology, Jinan University, Guangzhou, China
| | - Nobuko Akiyama
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Maki Miyauchi
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Hiroyuki Ohashi
- Division of Interactome Medical Sciences, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Etsuko Miyamoto-Sato
- Division of Interactome Medical Sciences, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki, Noda-shi, Chiba, Japan
| | - Hiroshi Yanagawa
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Taishin Akiyama
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Jun-ichiro Inoue
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| |
Collapse
|
41
|
Yamaguchi N, Yamaguchi N. The seventh zinc finger motif of A20 is required for the suppression of TNF-α-induced apoptosis. FEBS Lett 2015; 589:1369-75. [PMID: 25911380 DOI: 10.1016/j.febslet.2015.04.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 10/23/2022]
Abstract
The ubiquitin-editing enzyme A20 suppresses nuclear factor-κB (NF-κB) activation and tumor necrosis factor-α (TNF-α)-induced apoptosis in a deubiquitinating and ubiquitin ligase activity-dependent manner. Although recent studies revealed that A20 regulates NF-κB independently of its enzymatic activity through its seventh zinc finger motif (ZnF7), the involvement of ZnF7 in TNF-α-induced apoptosis is not clear. In this study, ZnF7 was found to be important for A20-mediated suppression of TNF-α-induced apoptosis. We also found that the ubiquitin ligases cIAP1/2 are required for A20 to suppress TNF-α-induced apoptosis. Because A20 binds to cIAP1/2 through ZnF7, these results suggest that A20 may control cIAP1/2 when suppressing TNF-α-induced apoptosis.
Collapse
Affiliation(s)
- Noritaka Yamaguchi
- Department of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan.
| | - Naoto Yamaguchi
- Department of Molecular Cell Biology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| |
Collapse
|
42
|
Korbolina EE, Ershov NI, Bryzgalov LO, Kolosova NG. Application of quantitative trait locus mapping and transcriptomics to studies of the senescence-accelerated phenotype in rats. BMC Genomics 2014; 15 Suppl 12:S3. [PMID: 25563673 PMCID: PMC4303943 DOI: 10.1186/1471-2164-15-s12-s3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Etiology of complex disorders, such as cataract and neurodegenerative diseases including age-related macular degeneration (AMD), remains poorly understood due to the paucity of animal models, fully replicating the human disease. Previously, two quantitative trait loci (QTLs) associated with early cataract, AMD-like retinopathy, and some behavioral aberrations in senescence-accelerated OXYS rats were uncovered on chromosome 1 in a cross between OXYS and WAG rats. To confirm the findings, we generated interval-specific congenic strains, WAG/OXYS-1.1 and WAG/OXYS-1.2, carrying OXYS-derived loci of chromosome 1 in the WAG strain. Both congenic strains displayed early cataract and retinopathy but differed clinically from OXYS rats. Here we applied a high-throughput RNA sequencing (RNA-Seq) strategy to facilitate nomination of the candidate genes and functional pathways that may be responsible for these differences and can contribute to the development of the senescence-accelerated phenotype of OXYS rats. Results First, the size and map position of QTL-derived congenic segments were determined by comparative analysis of coding single-nucleotide polymorphisms (SNPs), which were identified for OXYS, WAG, and congenic retinal RNAs after sequencing. The transferred locus was not what we expected in WAG/OXYS-1.1 rats. In rat retina, 15442 genes were expressed. Coherent sets of differentially expressed genes were identified when we compared RNA-Seq retinal profiles of 20-day-old WAG/OXYS-1.1, WAG/OXYS-1.2, and OXYS rats. The genes most different in the average expression level between the congenic strains included those generally associated with the Wnt, integrin, and TGF-β signaling pathways, widely involved in neurodegenerative processes. Several candidate genes (including Arhgap33, Cebpg, Gtf3c1, Snurf, Tnfaip3, Yme1l1, Cbs, Car9 and Fn1) were found to be either polymorphic in the congenic loci or differentially expressed between the strains. These genes may contribute to the development of cataract and retinopathy. Conclusions This study is the first RNA-Seq analysis of the rat retinal transcriptome generated with 40 mln sequencing read depth. The integration of QTL and transcriptomic analyses in our study forms the basis of future research into the relationship between the candidate genes within the congenic regions and specific changes in the retinal transcriptome as possible causal mechanisms that underlie age-associated disorders.
Collapse
|
43
|
Gu SY, Kim YE, Kwon KM, Han TH, Ahn JH. Biphasic regulation of A20 gene expression during human cytomegalovirus infection. Virol J 2014; 11:124. [PMID: 25005727 PMCID: PMC4104738 DOI: 10.1186/1743-422x-11-124] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/28/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The A20 ubiquitin-editing enzyme is a target of nuclear factor kappa B (NF-κB) and also plays a key role in regulating the NF-κB signaling pathway. NF-κB activity is increased during human cytomegalovirus (HCMV) infection and HCMV appears to be adapted to this change. To better understand the regulation of NF-κB signaling during HCMV infection, we investigated how A20 expression is controlled during HCMV infection. METHODS The expression level of A20 in human fibroblast cells infected with HCMV or UV-inactivated virus (UV-HCMV) was measured by immunoblot analysis, cell staining, and quantitative real-time PCR. Changes of histone modifications on the A20 promoter were determined by chromatin immunoprecipitation assays. Lentiviral vectors were used to knockdown A20 in fibroblast cells. RESULTS A20 expression was increased at early times after HCMV infection. This increase of the A20 protein level was promoted by viral gene expression under low viral load conditions. The viral IE1 protein, which is known to activate NF-κB, increased the A20 promoter activity through the upstream NF-κB sites in reporter assays, suggesting that IE1 is at least partly involved in A20 induction. Analysis of A20 expression with a high viral load demonstrated that the A20 regulation by HCMV was biphasic; both A20 protein and mRNA levels were increased at the early stage of infection, but decreased at the late stage. Under high viral load conditions, A20 upregulation was more profound with UV-HCMV than with HCMV, indicating a role of the viral gene product(s) in limiting A20 induction. Consistently, more histone modifications for euchromatin were found on the A20 promoter during UV-HCMV infection than with HCMV infection. A20 knockdown by shRNA reduced HCMV growth. CONCLUSION These results suggest that the biphasic regulation of A20 expression may be important for productive HCMV infection.
Collapse
Affiliation(s)
| | | | | | | | - Jin-Hyun Ahn
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 2066 Seoburo, Suwon 440-746, Republic of Korea.
| |
Collapse
|
44
|
Abstract
The NF-κB family of inducible transcription factors is activated in response to a variety of stimuli. Amongst the best-characterized inducers of NF-κB are members of the TNF family of cytokines. Research on NF-κB and TNF have been tightly intertwined for more than 25 years. Perhaps the most compelling examples of the interconnectedness of NF-κB and the TNF have come from analysis of knock-out mice that are unable to activate NF-κB. Such mice die embryonically, however, deletion of TNF or TNFR1 can rescue the lethality thereby illustrating the important role of NF-κB as the key regulator of transcriptional responses to TNF. The physiological connections between NF-κB and TNF cytokines are numerous and best explored in articles focusing on a single TNF family member. Instead, in this review, we explore general mechanisms of TNF cytokine signaling, with a focus on the upstream signaling events leading to activation of the so-called canonical and noncanonical NF-κB pathways by TNFR1 and CD40, respectively.
Collapse
Affiliation(s)
- Matthew S Hayden
- Department of Microbiology and Immunology, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA; Department of Dermatology, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA.
| | - Sankar Ghosh
- Department of Microbiology and Immunology, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA.
| |
Collapse
|
45
|
Guo S, Messmer-Blust AF, Wu J, Song X, Philbrick MJ, Shie JL, Rana JS, Li J. Role of A20 in cIAP-2 protection against tumor necrosis factor α (TNF-α)-mediated apoptosis in endothelial cells. Int J Mol Sci 2014; 15:3816-33. [PMID: 24595242 PMCID: PMC3975369 DOI: 10.3390/ijms15033816] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/30/2014] [Accepted: 02/06/2014] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor α (TNF-α) influences endothelial cell viability by altering the regulatory molecules involved in induction or suppression of apoptosis. However, the underlying mechanisms are still not completely understood. In this study, we demonstrated that A20 (also known as TNFAIP3, tumor necrosis factor α-induced protein 3, and an anti-apoptotic protein) regulates the inhibitor of apoptosis protein-2 (cIAP-2) expression upon TNF-α induction in endothelial cells. Inhibition of A20 expression by its siRNA resulted in attenuating expression of TNF-α-induced cIAP-2, yet not cIAP-1 or XIAP. A20-induced cIAP-2 expression can be blocked by the inhibition of phosphatidyl inositol-3 kinase (PI3-K), but not nuclear factor (NF)-κB, while concomitantly increasing the number of endothelial apoptotic cells and caspase 3 activation. Moreover, TNF-α-mediated induction of apoptosis was enhanced by A20 inhibition, which could be rescued by cIAP-2. Taken together, these results identify A20 as a cytoprotective factor involved in cIAP-2 inhibitory pathway of TNF-α-induced apoptosis. This is consistent with the idea that endothelial cell viability is dependent on interactions between inducers and suppressors of apoptosis, susceptible to modulation by TNF-α.
Collapse
Affiliation(s)
- Shuzhen Guo
- School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Angela F Messmer-Blust
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jiaping Wu
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Xiaoxiao Song
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Melissa J Philbrick
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jue-Lon Shie
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jamal S Rana
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jian Li
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
46
|
Enwere EK, Lacasse EC, Adam NJ, Korneluk RG. Role of the TWEAK-Fn14-cIAP1-NF-κB Signaling Axis in the Regulation of Myogenesis and Muscle Homeostasis. Front Immunol 2014; 5:34. [PMID: 24550918 PMCID: PMC3913901 DOI: 10.3389/fimmu.2014.00034] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/21/2014] [Indexed: 12/16/2022] Open
Abstract
Mammalian skeletal muscle maintains a robust regenerative capacity throughout life, largely due to the presence of a stem cell population known as “satellite cells” in the muscle milieu. In normal conditions, these cells remain quiescent; they are activated upon injury to become myoblasts, which proliferate extensively and eventually differentiate and fuse to form new multinucleated muscle fibers. Recent findings have identified some of the factors, including the cytokine TNFα-like weak inducer of apoptosis (TWEAK), which govern these cells’ decisions to proliferate, differentiate, or fuse. In this review, we will address the functions of TWEAK, its receptor Fn14, and the associated signal transduction molecule, the cellular inhibitor of apoptosis 1 (cIAP1), in the regulation of myogenesis. TWEAK signaling can activate the canonical NF-κB signaling pathway, which promotes myoblast proliferation and inhibits myogenesis. In addition, TWEAK activates the non-canonical NF-κB pathway, which, in contrast, promotes myogenesis by increasing myoblast fusion. Both pathways are regulated by cIAP1, which is an essential component of downstream signaling mediated by TWEAK and similar cytokines. This review will focus on the seemingly contradictory roles played by TWEAK during muscle regeneration, by highlighting the interplay between the two NF-κB pathways under physiological and pathological conditions. We will also discuss how myogenesis is negatively affected by chronic conditions, which affect homeostasis of the skeletal muscle environment.
Collapse
Affiliation(s)
- Emeka K Enwere
- Department of Medical Microbiology and Immunology, University of Alberta , Edmonton, AB , Canada
| | - Eric C Lacasse
- Solange Gauthier Karsh Molecular Genetics Laboratory, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute , Ottawa, ON , Canada
| | - Nadine J Adam
- Solange Gauthier Karsh Molecular Genetics Laboratory, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute , Ottawa, ON , Canada ; Department of Biochemistry, Microbiology and Immunology, University of Ottawa , Ottawa, ON , Canada
| | - Robert G Korneluk
- Solange Gauthier Karsh Molecular Genetics Laboratory, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute , Ottawa, ON , Canada ; Department of Biochemistry, Microbiology and Immunology, University of Ottawa , Ottawa, ON , Canada
| |
Collapse
|
47
|
Abstract
The ubiquitin system plays a pivotal role in the regulation of immune responses. This system includes a large family of E3 ubiquitin ligases of over 700 proteins and about 100 deubiquitinating enzymes, with the majority of their biological functions remaining unknown. Over the last decade, through a combination of genetic, biochemical, and molecular approaches, tremendous progress has been made in our understanding of how the process of protein ubiquitination and its reversal deubiquitination controls the basic aspect of the immune system including lymphocyte development, differentiation, activation, and tolerance induction and regulates the pathophysiological abnormalities such as autoimmunity, allergy, and malignant formation. In this review, we selected some of the published literature to discuss the roles of protein-ubiquitin conjugation and deubiquitination in T-cell activation and anergy, regulatory T-cell and T-helper cell differentiation, regulation of NF-κB signaling, and hematopoiesis in both normal and dysregulated conditions. A comprehensive understanding of the relationship between the ubiquitin system and immunity will provide insight into the molecular mechanisms of immune regulation and at the same time will advance new therapeutic intervention for human immunological diseases.
Collapse
Affiliation(s)
- Yoon Park
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Hyung-seung Jin
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Daisuke Aki
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Jeeho Lee
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Yun-Cai Liu
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA.
| |
Collapse
|
48
|
Vucic D. The Role of Ubiquitination in TWEAK-Stimulated Signaling. Front Immunol 2013; 4:472. [PMID: 24391645 PMCID: PMC3867686 DOI: 10.3389/fimmu.2013.00472] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 12/06/2013] [Indexed: 01/24/2023] Open
Abstract
Tumor necrosis factor superfamily ligands and receptors are responsible for development, immunity, and homeostasis of metazoan organisms. Thus, it is not surprising that signals emanating from these receptors are tightly regulated. Binding of TNF-related weak inducer of apoptosis (TWEAK) to its cognate receptor, FN14, triggers the assembly of receptor-associated signaling complex, which allows the activation of canonical and non-canonical nuclear factor kappa B (NF-κB) as well as mitogen-activated protein kinase signaling pathways. Ubiquitin ligases cellular inhibitor of apoptosis 1 and 2 (c-IAP1 and 2) and adaptor proteins TNFR-associated factors 2 and 3 (TRAF2 and TRAF3) are crucial for the regulation of TWEAK signaling as they facilitate the recruitment of distal signaling components including IKK and linear ubiquitin chain assembly complex complexes. At the same time c-IAP1/2, together with TRAF2 and TRAF3, promote constitutive ubiquitination and proteasomal degradation of NF-κB inducing kinase (NIK) – a kinase with critical role in the activation of non-canonical NF-κB signaling. While c-IAP1/2 mediated ubiquitination allows the activation of TWEAK-stimulated canonical NF-κB signaling, these E3 ligases are negative regulators of non-canonical signaling. TWEAK stimulation prompts the recruitment of c-IAP1/2 as well as TRAF2 and TRAF3 to the FN14 signaling complex leading to c-IAP1/2 autoubiquitination and degradation, which stabilizes NIK and allows subsequent phosphorylation of IKKα and partial proteasomal processing of p100 to activate gene expression. Recent studies have revealed that the spatio-temporal pattern of TWEAK-stimulated ubiquitination is a carefully orchestrated process involving several substrates that are modified by different ubiquitin linkages. Understanding the significance of ubiquitination for TWEAK signaling is important for the overall understanding of TWEAK biology and for the design of therapeutics that can be used in the treatment of human pathologies that are driven by TWEAK/FN14 expression and activity.
Collapse
Affiliation(s)
- Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, Inc. , South San Francisco, CA , USA
| |
Collapse
|