1
|
Glass AM, Navas-Martin S. Interferon-induced protein ISG15 in the central nervous system, quo vadis? FEBS Lett 2025. [PMID: 40353372 DOI: 10.1002/1873-3468.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/03/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025]
Abstract
The ubiquitin-like interferon (IFN)-stimulated gene 15 (ISG15) is a unique molecular effector that functions both intra- and extracellularly. Central to its pleiotropic nature is the ability to coordinate cellular responses following its conjugation to target proteins via ISGylation or in its free form. The activity of ISG15 is highly context-dependent: in the case of viral infections, ISG15 can serve as a pro- or antiviral factor. While ISG15 has been studied extensively, several gaps persist in our understanding of its role in dysregulated immune homeostasis. In particular, the role of ISG15 in the central nervous system (CNS), which has traditionally been considered an immune-privileged site, remains ill-defined. Interestingly, elevated ISG15 expression is observed in the CNS following instances of brain injury, autoimmunity, neurodegeneration, and viral infection. In this review, we seek to provide a comprehensive analysis of these studies as they pertain to ISG15 and its potential roles in the CNS. Furthermore, we discuss questions and challenges in the field while highlighting ISG15 as a potential diagnostic biomarker or therapeutic target. Impact statement While ISG15 has been studied extensively, several gaps remain in our understanding of its role in dysregulated immune homeostasis and its impact within the central nervous system (CNS). In this review, we provide a comprehensive analysis of the emerging roles of ISG15 in brain injury, autoimmunity, neurodegeneration, and viral infection within the CNS.
Collapse
Affiliation(s)
- Adam M Glass
- Department of Microbiology and Immunology, Centers for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Sonia Navas-Martin
- Department of Microbiology and Immunology, Centers for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
2
|
Liu Q, Wang X, Fang ZT, Zhao JN, Rui XX, Zhang BG, He Y, Liu RJ, Chen J, Chai GS, Liu GP. Upregulation of ISG15 induced by MAPT/tau accumulation represses autophagic flux by inhibiting HDAC6 activity: a vicious cycle in Alzheimer disease. Autophagy 2025; 21:807-826. [PMID: 39635882 PMCID: PMC11925114 DOI: 10.1080/15548627.2024.2431472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
Alzheimer disease (AD), a prevalent neurodegenerative condition in the elderly, is marked by a deficit in macroautophagy/autophagy, leading to intracellular MAPT/tau accumulation. While ISG15 (ISG15 ubiquitin like modifier) has been identified as a regulator of selective autophagy in ataxia telangiectasia (A-T), its role in AD remains unexplored. Our study reveals elevated ISG15 levels in the brains of patients with sporadic AD and AD models in vivo and in vitro. ISG15 overexpression in cells and the hippocampus inhibited HDAC6 (histone deacetylase 6) activity through C-terminal LRLRGG binding to HDAC6. Consequently, this increased CTTN (cortactin) acetylation, disrupted CTTN and F-actin recruitment to lysosomes, and impaired autophagosome (AP)-lysosome (LY) fusion. These disruptions led to MAPT/tau accumulation, synaptic damage, neuronal loss, and cognitive deficits. Conversely, ISG15 knockdown in our HsMAPT (human MAPT) pathology model restored HDAC6 activity, promoted AP-LY fusion, and improved cognitive function. This study identifies ISG15 as a key regulator of autophagic flux in AD, suggesting that targeting ISG15-mediated autophagy could offer therapeutic potential for AD.Abbreviation: AAV: adeno-associated virus; AD: Alzheimer disease; ALP: autophagy-lysosomal pathway; ANOVA: analysis of variance; AP: autophagosome; BafA1: bafilomycin A1; CHX: cycloheximide; CQ: chloroquine; CTTN: cortactin; FC: fear conditioning; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GRIN/NMDARs: N-methyl-D-aspartate glutamate ionotropic receptor NMDA types; HDAC6: histone deacetylase 6; HEK293: human embryonic kidney 293; HsMAPT: human MAPT; IF: immunofluorescence; IHC: immunohistochemistry; IP: immunoprecipitation; ISG15: ISG15 ubiquitin like modifier; LAMP1: lysosomal associated membrane protein 1; LY: lysosome; MAPT: microtubule associated protein tau; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MWM: Morris water maze; NOR: novel object recognition; SQSTM1/p62: sequestosome 1; ZnF UBP: zinc finger ubiquitin-binding protein.
Collapse
Affiliation(s)
- Qian Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Xin Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Ting Fang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun-Ning Zhao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Xiang Rui
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing-Ge Zhang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye He
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui-Juan Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gao-Shang Chai
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Department of Fundamental Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Subject of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
3
|
Davis GJ, Omole AO, Jung Y, Rut W, Holewinski R, Suazo KF, Kim HR, Yang M, Andresson T, Drag M, Yoo E. Chemical tools to define and manipulate interferon-inducible Ubl protease USP18. Nat Commun 2025; 16:957. [PMID: 39843430 PMCID: PMC11754618 DOI: 10.1038/s41467-025-56336-5] [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: 04/17/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
Ubiquitin-specific protease 18 (USP18) is a multifunctional cysteine protease primarily responsible for deconjugating the interferon-inducible ubiquitin-like modifier ISG15 from protein substrates. Here, we report the design and synthesis of activity-based probes (ABPs) that incorporate unnatural amino acids into the C-terminal tail of ISG15, enabling the selective detection of USP18 activity over other ISG15 cross-reactive deubiquitinases (DUBs) such as USP5 and USP14. Combined with a ubiquitin-based DUB ABP, the USP18 ABP is employed in a chemoproteomics screening platform to identify and assess inhibitors of DUBs including USP18. We further demonstrate that USP18 ABPs can be utilized to profile differential activities of USP18 in lung cancer cell lines, providing a strategy that will help define the activity-related landscape of USP18 in different disease states and unravel important (de)ISGylation-dependent biological processes.
Collapse
Affiliation(s)
- Griffin J Davis
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Anthony O Omole
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Yejin Jung
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Wioletta Rut
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Ronald Holewinski
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, USA
| | - Kiall F Suazo
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, USA
| | - Hong-Rae Kim
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Mo Yang
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Thorkell Andresson
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, USA
| | - Marcin Drag
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Euna Yoo
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| |
Collapse
|
4
|
Davis GJ, Omole AO, Jung Y, Rut W, Holewinski R, Suazo KF, Kim HR, Yang M, Andresson T, Drag M, Yoo E. Chemical tools to define and manipulate interferon-inducible Ubl protease USP18. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588544. [PMID: 38645224 PMCID: PMC11030383 DOI: 10.1101/2024.04.08.588544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Ubiquitin-specific protease 18 (USP18) is a multifunctional cysteine protease primarily responsible for deconjugating interferon-inducible ubiquitin-like (Ubl) modifier ISG15 from protein substrates. Here, we report the design and synthesis of activity-based probes (ABPs) capable of selectively detecting USP18 activity over other ISG15 cross-reactive deubiquitinases (DUBs) by incorporating unnatural amino acids into the C-terminal tail of ISG15. Combining with a ubiquitin-based DUB ABP, the selective USP18 ABP is employed in a chemoproteomic screening platform to identify and assess inhibitors of DUBs including USP18. We further demonstrate that USP18 ABPs can be utilized to profile differential activities of USP18 in lung cancer cell lines, providing a strategy that will help define the activity-related landscape of USP18 in different disease states and unravel important (de)ISGylation-dependent biological processes.
Collapse
Affiliation(s)
- Griffin J. Davis
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Anthony O. Omole
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Yejin Jung
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Wioletta Rut
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Ronald Holewinski
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland 21702, United States
| | - Kiall F. Suazo
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland 21702, United States
| | - Hong-Rae Kim
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
- Present address: Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02708, Korea
| | - Mo Yang
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Thorkell Andresson
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland 21702, United States
| | - Marcin Drag
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Euna Yoo
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| |
Collapse
|
5
|
Álvarez E, Falqui M, Sin L, McGrail JP, Perdiguero B, Coloma R, Marcos-Villar L, Tárrega C, Esteban M, Gómez CE, Guerra S. Unveiling the Multifaceted Roles of ISG15: From Immunomodulation to Therapeutic Frontiers. Vaccines (Basel) 2024; 12:153. [PMID: 38400136 PMCID: PMC10891536 DOI: 10.3390/vaccines12020153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The Interferon Stimulated Gene 15 (ISG15), a unique Ubiquitin-like (Ubl) modifier exclusive to vertebrates, plays a crucial role in the immune system. Primarily induced by interferon (IFN) type I, ISG15 functions through diverse mechanisms: (i) covalent protein modification (ISGylation); (ii) non-covalent intracellular action; and (iii) exerting extracellular cytokine activity. These various roles highlight its versatility in influencing numerous cellular pathways, encompassing DNA damage response, autophagy, antiviral response, and cancer-related processes, among others. The well-established antiviral effects of ISGylation contrast with its intriguing dual role in cancer, exhibiting both suppressive and promoting effects depending on the tumour type. The multifaceted functions of ISG15 extend beyond intracellular processes to extracellular cytokine signalling, influencing immune response, chemotaxis, and anti-tumour effects. Moreover, ISG15 emerges as a promising adjuvant in vaccine development, enhancing immune responses against viral antigens and demonstrating efficacy in cancer models. As a therapeutic target in cancer treatment, ISG15 exhibits a double-edged nature, promoting or suppressing oncogenesis depending on the tumour context. This review aims to contribute to future studies exploring the role of ISG15 in immune modulation and cancer therapy, potentially paving the way for the development of novel therapeutic interventions, vaccine development, and precision medicine.
Collapse
Affiliation(s)
- Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
| | - Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
| | - Laura Sin
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Joseph Patrick McGrail
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Rocío Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Céline Tárrega
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.S.); (B.P.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.F.); (J.P.M.); (R.C.); (C.T.)
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
6
|
Sarkar R, Patra U, Mukherjee A, Mitra S, Komoto S, Chawla-Sarkar M. Rotavirus circumvents the antiviral effects of protein ISGylation via proteasomal degradation of Ube1L. Cell Signal 2023; 112:110891. [PMID: 37722521 DOI: 10.1016/j.cellsig.2023.110891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/10/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Among the ramified cellular responses elicited in response to pathogenic stimuli, upregulation and covalent conjugation of an Ubiquitin-like modifier ISG15 to lysine residues of target proteins (ISGylation) through sequential action of three enzymes E1 (Ube1L), E2 (Ube2L6) and E3 (Herc5) have emerged as an important regulatory facet governing innate immunity against numerous viral infections. In the present study, we investigated the interplay between host ISGylation system and Rotavirus (RV). We observed that RV infection upregulates the expression of free ISG15 but prevents protein ISGylation. Analysing the expression of ISGylation machinery components revealed that RV infection results in steady depletion of Ube1L protein with the progression of infection. Indeed, restoration of Ube1L expression caused induction in protein ISGylation during RV infection. Subsequent investigation revealed that ectopic expression of RV non-structural protein 5 (NSP5) fosters proteolytic ubiquitylation of Ube1L, thereby depleting it in an ubiquitin-proteasome-dependent manner. Moreover, pan-Cullin inhibition also abrogates proteolytic ubiquitylation and rescued depleted Ube1L in RV-NSP5 expressing cells, suggesting the involvement of host cellular Cullin RING Ligases (CRLs) in proteasomal degradation of Ube1L during RV-SA11 infection. Reciprocal co-immunoprecipitation analyses substantiated a molecular association between Ube1L and RV-NSP5 during infection scenario and also under ectopically overexpressed condition independent of intermediate RNA scaffold and RV-NSP5 hyperphosphorylation. Interestingly, clonal overexpression of Ube1L reduced expression of RV proteins and RV infectivity, which are restored in ISG15 silenced cells, suggesting that Ube1L is a crucial anti-viral host cellular determinant that inhibits RV infection by promoting the formation of ISG15 conjugates.
Collapse
Affiliation(s)
- Rakesh Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Upayan Patra
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Arpita Mukherjee
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Suvrotoa Mitra
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Satoshi Komoto
- Department of Virology and Parasitology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Mamta Chawla-Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India.
| |
Collapse
|
7
|
Falqui M, Perdiguero B, Coloma R, Albert M, Marcos-Villar L, McGrail JP, Sorzano CÓS, Esteban M, Gómez CE, Guerra S. An MVA-based vector expressing cell-free ISG15 increases IFN-I production and improves HIV-1-specific CD8 T cell immune responses. Front Cell Infect Microbiol 2023; 13:1187193. [PMID: 37313341 PMCID: PMC10258332 DOI: 10.3389/fcimb.2023.1187193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/17/2023] [Indexed: 06/15/2023] Open
Abstract
The human immunodeficiency virus (HIV), responsible of the Acquired Immune Deficiency Syndrome (AIDS), continues to be a major global public health issue with any cure or vaccine available. The Interferon-stimulated gene 15 (ISG15) encodes a ubiquitin-like protein that is induced by interferons and plays a critical role in the immune response. ISG15 is a modifier protein that covalently binds to its targets via a reversible bond, a process known as ISGylation, which is the best-characterized activity of this protein to date. However, ISG15 can also interact with intracellular proteins via non-covalent binding or act as a cytokine in the extracellular space after secretion. In previous studies we proved the adjuvant effect of ISG15 when delivered by a DNA-vector in heterologous prime-boost combination with a Modified Vaccinia virus Ankara (MVA)-based recombinant virus expressing HIV-1 antigens Env/Gag-Pol-Nef (MVA-B). Here we extended these results evaluating the adjuvant effect of ISG15 when expressed by an MVA vector. For this, we generated and characterized two novel MVA recombinants expressing different forms of ISG15, the wild-type ISG15GG (able to perform ISGylation) or the mutated ISG15AA (unable to perform ISGylation). In mice immunized with the heterologous DNA prime/MVA boost regimen, the expression of the mutant ISG15AA from MVA-Δ3-ISG15AA vector in combination with MVA-B induced an increase in the magnitude and quality of HIV-1-specific CD8 T cells as well as in the levels of IFN-I released, providing a better immunostimulatory activity than the wild-type ISG15GG. Our results confirm the importance of ISG15 as an immune adjuvant in the vaccine field and highlights its role as a potential relevant component in HIV-1 immunization protocols.
Collapse
Affiliation(s)
- Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rocio Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Albert
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Joseph Patrick McGrail
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Óscar S. Sorzano
- Biocomputing Unit and Computational Genomics, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
8
|
Nguyen HM, Gaikwad S, Oladejo M, Agrawal MY, Srivastava SK, Wood LM. Interferon stimulated gene 15 (ISG15) in cancer: An update. Cancer Lett 2023; 556:216080. [PMID: 36736853 DOI: 10.1016/j.canlet.2023.216080] [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: 11/16/2022] [Revised: 01/17/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023]
Abstract
Among the plethora of defense mechanisms which a host elicits after pathogen invasion, type 1 interferons play a central role in regulating the immune system's response. They induce several interferon-stimulated genes (ISGs) which play a diverse role once activated. Over the past few decades, there have been several studies exploring the role of ISGs in cancer and ISG15 is among the most studied for its pro and anti-tumorigenic role. In this review, we aim to provide an update on the recent observations and findings related to ISG15 in cancer. We provide a brief overview about the initial observations and important historical findings which helped scientists understand structure and function of ISG15. We aim to provide an overview of ISG15 in cancer with an emphasis on studies which delve into the molecular mechanism of ISG15 in modulating the tumor microenvironment. Further, the dysregulation of ISG15 in cancer and the molecular mechanisms associated with its pro and anti-tumor roles are discussed in respective cancer types. Finally, we discuss multiple therapeutic applications of ISG15 in current cancer therapy.
Collapse
Affiliation(s)
- Hong-My Nguyen
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Shreyas Gaikwad
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Manas Yogendra Agrawal
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Sanjay K Srivastava
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Laurence M Wood
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA.
| |
Collapse
|
9
|
Coronaviral PLpro proteases and the immunomodulatory roles of conjugated versus free Interferon Stimulated Gene product-15 (ISG15). Semin Cell Dev Biol 2022; 132:16-26. [PMID: 35764457 PMCID: PMC9233553 DOI: 10.1016/j.semcdb.2022.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 06/12/2022] [Accepted: 06/13/2022] [Indexed: 12/14/2022]
Abstract
Ubiquitin-like proteins (Ubls) share some features with ubiquitin (Ub) such as their globular 3D structure and the ability to attach covalently to other proteins. Interferon Stimulated Gene 15 (ISG15) is an abundant Ubl that similar to Ub, marks many hundreds of cellular proteins, altering their fate. In contrast to Ub, , ISG15 requires interferon (IFN) induction to conjugate efficiently to other proteins. Moreover, despite the multitude of E3 ligases for Ub-modified targets, a single E3 ligase termed HERC5 (in humans) is responsible for the bulk of ISG15 conjugation. Targets include both viral and cellular proteins spanning an array of cellular compartments and metabolic pathways. So far, no common structural or biochemical feature has been attributed to these diverse substrates, raising questions about how and why they are selected. Conjugation of ISG15 mitigates some viral and bacterial infections and is linked to a lower viral load pointing to the role of ISG15 in the cellular immune response. In an apparent attempt to evade the immune response, some viruses try to interfere with the ISG15 pathway. For example, deconjugation of ISG15 appears to be an approach taken by coronaviruses to interfere with ISG15 conjugates. Specifically, coronaviruses such as SARS-CoV, MERS-CoV, and SARS-CoV-2, encode papain-like proteases (PL1pro) that bear striking structural and catalytic similarities to the catalytic core domain of eukaryotic deubiquitinating enzymes of the Ubiquitin-Specific Protease (USP) sub-family. The cleavage specificity of these PLpro enzymes is for flexible polypeptides containing a consensus sequence (R/K)LXGG, enabling them to function on two seemingly unrelated categories of substrates: (i) the viral polyprotein 1 (PP1a, PP1ab) and (ii) Ub- or ISG15-conjugates. As a result, PLpro enzymes process the viral polyprotein 1 into an array of functional proteins for viral replication (termed non-structural proteins; NSPs), and it can remove Ub or ISG15 units from conjugates. However, by de-conjugating ISG15, the virus also creates free ISG15, which in turn may affect the immune response in two opposite pathways: free ISG15 negatively regulates IFN signaling in humans by binding non-catalytically to USP18, yet at the same time free ISG15 can be secreted from the cell and induce the IFN pathway of the neighboring cells. A deeper understanding of this protein-modification pathway and the mechanisms of the enzymes that counteract it will bring about effective clinical strategies related to viral and bacterial infections.
Collapse
|
10
|
Schwartzenburg J, Reed R, Koul H, Zea AH, Shellito J, Miele L, Crabtree JS, Desai S. ISGylation is increased in the peripheral blood mononuclear cells derived from symptomatic COVID-19 patients. Exp Biol Med (Maywood) 2022; 247:842-847. [PMID: 35130743 DOI: 10.1177/15353702221075606] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cytokine-driven hyper inflammation has been identified as a critical factor behind poor outcomes in patients severely infected with SARS-CoV-2 virus. Notably, protein ISGylation, a protein conjugated form of Type 1 IFN-inducible ubiquitin-like protein ISG15 (Interferon-Stimulated Gene 15), induces cytokine storm (CS) and augments colonic inflammation in colitis-associated colon cancers in mouse models. However, whether ISGylation is increased and causally responsible for CS and hyper inflammation in symptomatic COVID-19 patients is unknown. Here, we measured ISGylation levels in peripheral blood mononuclear cells (PBMCs) from 10 symptomatic (SARS-CoV-2-positive with symptoms) and asymptomatic (SARS-CoV-2-positive with no symptoms) COVID-19 patients, and 4 uninfected individuals (SARS-CoV-2-negative), using WesTm assay. Strikingly, we note significant increases in protein ISGylation and MX-1 (myxovirus-resistance protein-1) protein levels, both induced by type-I IFN, in symptomatic but not in asymptomatic patients and uninfected individuals. Knowing that ISGylation augments CS and intestinal inflammation in colon cancers, we propose that increased ISGylation may be an underlying cause of CS and inflammation in symptomatic patients.
Collapse
Affiliation(s)
- Joshua Schwartzenburg
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-School of Medicine, New Orleans, LA 70112, USA
| | - Ryan Reed
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-School of Medicine, New Orleans, LA 70112, USA
| | - Hari Koul
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-School of Medicine, New Orleans, LA 70112, USA
| | - Arnold H Zea
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center-School of Medicine, New Orleans, LA 70112, USA
| | - Judd Shellito
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center-School of Medicine, New Orleans, LA 70112, USA
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center-School of Medicine, New Orleans, LA 70112, USA
| | - Judy S Crabtree
- Department of Genetics, Louisiana State University Health Sciences Center-School of Medicine, New Orleans, LA 70112, USA
| | - Shyamal Desai
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
11
|
Mirzalieva O, Juncker M, Schwartzenburg J, Desai S. ISG15 and ISGylation in Human Diseases. Cells 2022; 11:cells11030538. [PMID: 35159348 PMCID: PMC8834048 DOI: 10.3390/cells11030538] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 12/04/2022] Open
Abstract
Type I Interferons (IFNs) induce the expression of >500 genes, which are collectively called ISGs (IFN-stimulated genes). One of the earliest ISGs induced by IFNs is ISG15 (Interferon-Stimulated Gene 15). Free ISG15 protein synthesized from the ISG15 gene is post-translationally conjugated to cellular proteins and is also secreted by cells into the extracellular milieu. ISG15 comprises two ubiquitin-like domains (UBL1 and UBL2), each of which bears a striking similarity to ubiquitin, accounting for its earlier name ubiquitin cross-reactive protein (UCRP). Like ubiquitin, ISG15 harbors a characteristic β-grasp fold in both UBL domains. UBL2 domain has a conserved C-terminal Gly-Gly motif through which cellular proteins are appended via an enzymatic cascade similar to ubiquitylation called ISGylation. ISG15 protein is minimally expressed under physiological conditions. However, its IFN-dependent expression is aberrantly elevated or compromised in various human diseases, including multiple types of cancer, neurodegenerative disorders (Ataxia Telangiectasia and Amyotrophic Lateral Sclerosis), inflammatory diseases (Mendelian Susceptibility to Mycobacterial Disease (MSMD), bacteriopathy and viropathy), and in the lumbar spinal cords of veterans exposed to Traumatic Brain Injury (TBI). ISG15 and ISGylation have both inhibitory and/or stimulatory roles in the etiology and pathogenesis of human diseases. Thus, ISG15 is considered a “double-edged sword” for human diseases in which its expression is elevated. Because of the roles of ISG15 and ISGylation in cancer cell proliferation, migration, and metastasis, conferring anti-cancer drug sensitivity to tumor cells, and its elevated expression in cancer, neurodegenerative disorders, and veterans exposed to TBI, both ISG15 and ISGylation are now considered diagnostic/prognostic biomarkers and therapeutic targets for these ailments. In the current review, we shall cover the exciting journey of ISG15, spanning three decades from the bench to the bedside.
Collapse
Affiliation(s)
| | | | | | - Shyamal Desai
- Correspondence: ; Tel.: +1-504-568-4388; Fax: +1-504-568-2093
| |
Collapse
|
12
|
Juncker M, Kim C, Reed R, Haas A, Schwartzenburg J, Desai S. ISG15 attenuates post-translational modifications of mitofusins and congression of damaged mitochondria in Ataxia Telangiectasia cells. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166102. [PMID: 33617986 DOI: 10.1016/j.bbadis.2021.166102] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 10/22/2022]
Abstract
Mitophagy is defective in several neurodegenerative diseases, including Ataxia Telangiectasia (A-T). However, the molecular mechanism underlying defective mitophagy in A-T is unknown. Literature indicates that damaged mitochondria are transported to the perinuclear region prior to their removal via mitophagy. Our previous work has indicated that conjugation of SUMO2 (Small Ubiquitin-like Modifier 2) to mitofusins (Mfns) may be necessary for congression of mitochondria into SUMO2-/ubiquitin-/LC3-positive compact structures resembling mito-aggresomes at the perinuclear region in CCCP-treated HEK293 cells. Here, we demonstrate that Mfns are SUMOylated, and mitochondria are transported to the perinuclear region; however, mitochondria fail to congress into mito-aggresome-like structures in CCCP-treated A-T cells. Defect in mitochondrial congression is causally related to constitutively elevated ISG15 (Interferon-Stimulated Gene 15), an antagonist of the ubiquitin pathway, in A-T cells. Suppression of the ISG15 pathway restores mitochondrial congression, reduce oxidative stress, and level of unhealthy mitochondria, which is suggestive of restoration of mitophagy in A-T cells. ISG15 is also constitutively elevated and mitophagy is defective in Amytrophic Lateral Sclerosis (ALS). The constitutively elevated ISG15 pathway therefore appears to be a common unifying biochemical mechanism underlying defective mitophagy in neurodegenerative disorders thus, implying the broader significance of our findings, and suggest the potential role of ISG15 inhibitors in their treatment.
Collapse
Affiliation(s)
- Meredith Juncker
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, 1901 Perdido Street, New Orleans, LA 70112, USA
| | - Catherine Kim
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, 1901 Perdido Street, New Orleans, LA 70112, USA
| | - Ryan Reed
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, 1901 Perdido Street, New Orleans, LA 70112, USA
| | - Arthur Haas
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, 1901 Perdido Street, New Orleans, LA 70112, USA
| | - Joshua Schwartzenburg
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, 1901 Perdido Street, New Orleans, LA 70112, USA
| | - Shyamal Desai
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, 1901 Perdido Street, New Orleans, LA 70112, USA.
| |
Collapse
|
13
|
Chelbi-Alix MK, Thibault P. Crosstalk Between SUMO and Ubiquitin-Like Proteins: Implication for Antiviral Defense. Front Cell Dev Biol 2021; 9:671067. [PMID: 33968942 PMCID: PMC8097047 DOI: 10.3389/fcell.2021.671067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Interferon (IFN) is a crucial first line of defense against viral infection. This cytokine induces the expression of several IFN-Stimulated Genes (ISGs), some of which act as restriction factors. Upon IFN stimulation, cells also express ISG15 and SUMO, two key ubiquitin-like (Ubl) modifiers that play important roles in the antiviral response. IFN itself increases the global cellular SUMOylation in a PML-dependent manner. Mass spectrometry-based proteomics enables the large-scale identification of Ubl protein conjugates to determine the sites of modification and the quantitative changes in protein abundance. Importantly, a key difference amongst SUMO paralogs is the ability of SUMO2/3 to form poly-SUMO chains that recruit SUMO ubiquitin ligases such RING finger protein RNF4 and RNF111, thus resulting in the proteasomal degradation of conjugated substrates. Crosstalk between poly-SUMOylation and ISG15 has been reported recently, where increased poly-SUMOylation in response to IFN enhances IFN-induced ISGylation, stabilizes several ISG products in a TRIM25-dependent fashion, and results in enhanced IFN-induced antiviral activities. This contribution will highlight the relevance of the global SUMO proteome and the crosstalk between SUMO, ubiquitin and ISG15 in controlling both the stability and function of specific restriction factors that mediate IFN antiviral defense.
Collapse
Affiliation(s)
| | - Pierre Thibault
- Institute for Research in Immunology and Cancer, Montréal, QC, Canada
- Department of Chemistry, University of Montreal, Montréal, QC, Canada
| |
Collapse
|
14
|
Ford BD, Moncada Giraldo D, Margaroli C, Giacalone VD, Brown MR, Peng L, Tirouvanziam R. Functional and Transcriptional Adaptations of Blood Monocytes Recruited to the Cystic Fibrosis Airway Microenvironment In Vitro. Int J Mol Sci 2021; 22:2530. [PMID: 33802410 PMCID: PMC7959310 DOI: 10.3390/ijms22052530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/17/2022] Open
Abstract
Cystic fibrosis (CF) lung disease is dominated by the recruitment of myeloid cells (neutrophils and monocytes) from the blood which fail to clear the lung of colonizing microbes. In prior in vitro studies, we showed that blood neutrophils migrated through the well-differentiated lung epithelium into the CF airway fluid supernatant (ASN) mimic the dysfunction of CF airway neutrophils in vivo, including decreased bactericidal activity despite an increased metabolism. Here, we hypothesized that, in a similar manner to neutrophils, blood monocytes undergo significant adaptations upon recruitment to CFASN. To test this hypothesis, primary human blood monocytes were transmigrated in our in vitro model into the ASN from healthy control (HC) or CF subjects to mimic in vivo recruitment to normal or CF airways, respectively. Surface phenotype, metabolic and bacterial killing activities, and transcriptomic profile by RNA sequencing were quantified post-transmigration. Unlike neutrophils, monocytes were not metabolically activated, nor did they show broad differences in activation and scavenger receptor expression upon recruitment to the CFASN compared to HCASN. However, monocytes recruited to CFASN showed decreased bactericidal activity. RNASeq analysis showed strong effects of transmigration on monocyte RNA profile, with differences between CFASN and HCASN conditions, notably in immune signaling, including lower expression in the former of the antimicrobial factor ISG15, defensin-like chemokine CXCL11, and nitric oxide-producing enzyme NOS3. While monocytes undergo qualitatively different adaptations from those seen in neutrophils upon recruitment to the CF airway microenvironment, their bactericidal activity is also dysregulated, which could explain why they also fail to protect CF airways from infection.
Collapse
Affiliation(s)
- Bijean D. Ford
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (B.D.F.); (D.M.G.); (V.D.G.); (M.R.B.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Diego Moncada Giraldo
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (B.D.F.); (D.M.G.); (V.D.G.); (M.R.B.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Camilla Margaroli
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Vincent D. Giacalone
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (B.D.F.); (D.M.G.); (V.D.G.); (M.R.B.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Milton R. Brown
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (B.D.F.); (D.M.G.); (V.D.G.); (M.R.B.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Limin Peng
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA 30322, USA;
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (B.D.F.); (D.M.G.); (V.D.G.); (M.R.B.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| |
Collapse
|
15
|
Wu SF, Xia L, Shi XD, Dai YJ, Zhang WN, Zhao JM, Zhang W, Weng XQ, Lu J, Le HY, Tao SC, Zhu J, Chen Z, Wang YY, Chen S. RIG-I regulates myeloid differentiation by promoting TRIM25-mediated ISGylation. Proc Natl Acad Sci U S A 2020; 117:14395-14404. [PMID: 32513696 PMCID: PMC7322067 DOI: 10.1073/pnas.1918596117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Retinoic acid-inducible gene I (RIG-I) is up-regulated during granulocytic differentiation of acute promyelocytic leukemia (APL) cells induced by all-trans retinoic acid (ATRA). It has been reported that RIG-I recognizes virus-specific 5'-ppp-double-stranded RNA (dsRNA) and activates the type I interferons signaling pathways in innate immunity. However, the functions of RIG-I in hematopoiesis remain unclear, especially regarding its possible interaction with endogenous RNAs and the associated pathways that could contribute to the cellular differentiation and maturation. Herein, we identified a number of RIG-I-binding endogenous RNAs in APL cells following ATRA treatment, including the tripartite motif-containing protein 25 (TRIM25) messenger RNA (mRNA). TRIM25 encodes the protein known as an E3 ligase for ubiquitin/interferon (IFN)-induced 15-kDa protein (ISG15) that is involved in RIG-I-mediated antiviral signaling. We show that RIG-I could bind TRIM25 mRNA via its helicase domain and C-terminal regulatory domain, enhancing the stability of TRIM25 transcripts. RIG-I could increase the transcriptional expression of TRIM25 by caspase recruitment domain (CARD) domain through an IFN-stimulated response element. In addition, RIG-I activated other key genes in the ISGylation pathway by activating signal transducer and activator of transcription 1 (STAT1), including the modifier ISG15 and several enzymes responsible for the conjugation of ISG15 to protein substrates. RIG-I cooperated with STAT1/2 and interferon regulatory factor 1 (IRF1) to promote the activation of the ISGylation pathway. The integrity of ISGylation in ATRA or RIG-I-induced cell differentiation was essential given that knockdown of TRIM25 or ISG15 resulted in significant inhibition of this process. Our results provide insight into the role of the RIG-I-TRIM25-ISGylation axis in myeloid differentiation.
Collapse
Affiliation(s)
- Song-Fang Wu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Li Xia
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Dong Shi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu-Jun Dai
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei-Na Zhang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jun-Mei Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wu Zhang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiang-Qin Weng
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Lu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huang-Ying Le
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Sheng-Ce Tao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiang Zhu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhu Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yue-Ying Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Saijuan Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Rui Jin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
16
|
Carriere J, Rao Y, Liu Q, Lin X, Zhao J, Feng P. Post-translational Control of Innate Immune Signaling Pathways by Herpesviruses. Front Microbiol 2019; 10:2647. [PMID: 31798565 PMCID: PMC6868034 DOI: 10.3389/fmicb.2019.02647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/30/2019] [Indexed: 12/21/2022] Open
Abstract
Herpesviruses constitute a large family of disease-causing DNA viruses. Each herpesvirus strain is capable of infecting particular organisms with a specific cell tropism. Upon infection, pattern recognition receptors (PRRs) recognize conserved viral features to trigger signaling cascades that culminate in the production of interferons and pro-inflammatory cytokines. To invoke a proper immune response while avoiding collateral tissue damage, signaling proteins involved in these cascades are tightly regulated by post-translational modifications (PTMs). Herpesviruses have developed strategies to subvert innate immune signaling pathways in order to ensure efficient viral replication and achieve persistent infection. The ability of these viruses to control the proteins involved in these signaling cascades post-translationally, either directly via virus-encoded enzymes or indirectly through the deregulation of cellular enzymes, has been widely reported. This ability provides herpesviruses with a powerful tool to shut off or restrict host antiviral and inflammatory responses. In this review, we highlight recent findings on the herpesvirus-mediated post-translational control along PRR-mediated signaling pathways.
Collapse
Affiliation(s)
| | | | | | | | | | - Pinghui Feng
- Section of Infection and Immunity, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
17
|
Chandrakar K, Jain A, Khan JR, Jain T, Singh M, Mishra OP. Molecular characterization and expression profile of interferon-stimulated gene 15 (ISG15) in the endometrium of goat (Capra hircus). Theriogenology 2019; 142:348-354. [PMID: 31711698 DOI: 10.1016/j.theriogenology.2019.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 09/17/2019] [Accepted: 10/13/2019] [Indexed: 11/28/2022]
Abstract
Interferon-stimulated gene 15 (ISG15), a ubiquitin-like protein, is responsible for uterine receptivity, implantation and conceptus development in different ruminant species, but in goat (Capra hircus) its role is yet to be explicated. In the present study, the ISG15 gene was cloned, characterized and its temporal expression profile was examined in the endometrium of caprine (cp). A fragment of cpISG15 gene, 1033 bp in length, was amplified, cloned and sequenced from genomic DNA covering the coding region. Sequence analysis of cpISG15 gene revealed that it was comprised of two exons of 59 bp and 496 bp encoding a peptide of 157 amino acids. Complementary DNA (cDNA) and deduced amino acid sequences of cpISG15 exhibited 99 and 98, 93 and 88, 94 and 89, 76 and 66, and 73 and 62% identity with that of sheep, cattle, buffalo, human and mice, respectively. Further, relative expression of cpISG15 mRNA and protein was determined by quantitative real-time PCR (qPCR) and Western blot, respectively, in the endometrium of pregnant and cyclic does. Both cpISG15 mRNA and protein were expressed maximally (P < 0.05) in the endometrium during early stage of pregnancy (16-24 d) as compared to cyclic does, but no significant difference was observed in cpISG15 mRNA and protein expression in the endometrium between the later stage of pregnancy (25-40 d) and cyclic does. It is concluded that cpISG15 is almost similar in structure and probably in function also to other species as it has been found significantly upregulated during early pregnancy.
Collapse
Affiliation(s)
- K Chandrakar
- Veterinary Assistant Surgeon, Govt. of Chhattisgarh, Mahasamund, Chhattisgarh, India
| | - A Jain
- Molecular Genetics Laboratory, Department of Animal Genetics and Breeding, College of Veterinary Science and Animal Husbandry, CGKV, Anjora, Durg, Chhattisgarh, India.
| | - J R Khan
- Department of Veterinary Physiology and Biochemistry, College of Veterinary Science and Animal Husbandry, CGKV, Anjora, Durg, Chhattisgarh, India
| | - T Jain
- Animal Biotechnology Centre, CGKV, Durg, Chhattisgarh, India
| | - M Singh
- Molecular Genetics Laboratory, Department of Animal Genetics and Breeding, College of Veterinary Science and Animal Husbandry, CGKV, Anjora, Durg, Chhattisgarh, India
| | - O P Mishra
- Department of Veterinary Physiology and Biochemistry, College of Veterinary Science and Animal Husbandry, CGKV, Anjora, Durg, Chhattisgarh, India
| |
Collapse
|
18
|
Abstract
The host response to viral infection includes the induction of type I interferons and the subsequent upregulation of hundreds of interferon-stimulated genes. Ubiquitin-like protein ISG15 is an interferon-induced protein that has been implicated as a central player in the host antiviral response. Over the past 15 years, efforts to understand how ISG15 protects the host during infection have revealed that its actions are diverse and pathogen-dependent. In this Review, we describe new insights into how ISG15 directly inhibits viral replication and discuss the recent finding that ISG15 modulates the host damage and repair response, immune response and other host signalling pathways. We also explore the viral immune-evasion strategies that counteract the actions of ISG15. These findings are integrated with a discussion of the recent identification of ISG15-deficient individuals and a cellular receptor for ISG15 that provides new insights into how ISG15 shapes the host response to viral infection. Ubiquitin-like protein ISG15 is an interferon-induced protein that has been implicated as a central player in the host antiviral response. In this Review, Perng and Lenschow provide new insights into how ISG15 restricts and shapes the host response to viral infection and the viral immune-evasion strategies that counteract ISG15.
Collapse
Affiliation(s)
- Yi-Chieh Perng
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Deborah J Lenschow
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA. .,Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
19
|
Schwartzenburg J, Juncker M, Reed R, Desai S. Increased ISGylation in Cases of TBI-Exposed ALS Veterans. J Neuropathol Exp Neurol 2019; 78:209-218. [PMID: 30657969 PMCID: PMC6380302 DOI: 10.1093/jnen/nly129] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Veterans who have served in the military are at a nearly 60% greater risk of being diagnosed with amyotrophic lateral sclerosis (ALS). Literature reports suggest that a history of traumatic brain injury (TBI) may be a risk factor for ALS in veterans. However, no diagnostic biomarkers are available for identifying ALS risk/development in TBI-exposed veterans. Here, using a Wes assay, we show that ISGylation, a conjugated form of interferon-stimulated gene 15 protein, is significantly elevated in the lumbar spinal cords (SC-Ls) of TBI-ALS compared with ALS veterans without a previous history of TBI (nonTBI-ALS). Although not as striking as in TBI-ALS veterans, ISGylation is also increased in nonTBI-ALS compared with normal veterans. Notably, no changes in ISGylation were seen in occipital lobe samples obtained from the same patients, suggesting that elevated ISGylation is distinct to ALS disease-specific SC-Ls. Moreover, we detected increased ISGylation in cerebral spinal fluid samples of TBI-ALS veterans. Other results using cultured lymphocyte cell lines show a similar trend of increased ISGylation in ALS patients from the general population. Together, these data suggest that ISGylation could serve as a diagnostic biomarker for TBI-ALS veterans, nonTBI-ALS veterans, and nonveterans affected by ALS.
Collapse
Affiliation(s)
- Joshua Schwartzenburg
- Department of Biochemistry and Molecular Biology, LSUHSC-School of Medicine, New Orleans, Louisiana
| | - Meredith Juncker
- Department of Biochemistry and Molecular Biology, LSUHSC-School of Medicine, New Orleans, Louisiana
| | - Ryan Reed
- Department of Biochemistry and Molecular Biology, LSUHSC-School of Medicine, New Orleans, Louisiana
| | - Shyamal Desai
- Department of Biochemistry and Molecular Biology, LSUHSC-School of Medicine, New Orleans, Louisiana
| |
Collapse
|
20
|
Inflammation-dependent ISG15 upregulation mediates MIA-induced dendrite damages and depression by disrupting NEDD4/Rap2A signaling. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1477-1489. [PMID: 30826466 DOI: 10.1016/j.bbadis.2019.02.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Maternal immune activation (MIA) is an independent risk factor for psychiatric disorders including depression spectrum in the offsprings, but the molecular mechanism is unclear. Recent studies show that interferon-stimulated gene-15 (ISG15) is involved in inflammation and neuronal dendrite development; here we studied the role of ISG15 in MIA-induced depression and the underlying mechanisms. METHODS By vena caudalis injecting polyinosinic: polycytidylic acid (poly I:C) into the pregnant rats to mimic MIA, we used AAV or lentivirus to introduce or silence ISG15 expression. Synaptic plasticity was detected by confocal microscope and Golgi staining. Cognitive performances of the offspring were measured by Open field, Forced swimming and Sucrose preference test. RESULTS We found that MIA induced depression-like behaviors with dendrite impairments in the offspring with ISG15 level increased in the offsprings' brain. Overexpressing ISG15 in the prefrontal cortex of neonatal cubs (P0) could mimic dendritic pathology and depressive like behaviors, while downregulating ISG15 rescued these abnormalities in the offsprings. Further studies demonstrated that MIA-induced upregulation of inflammatory cytokines promoted ISG15 expression in the offspring' brain which suppressed Rap2A ubiquitination via NEDD4 and thus induced Rap2A accumulation, while upregulating NEDD4 abolished ISG15-induced dendrite impairments. CONCLUSIONS These data reveal that MIA impedes offsprings' dendrite development and causes depressive like behaviors by upregulating ISG15 and suppressing NEDD4/Rap2A signaling. The current findings suggest that inhibiting ISG15 may be a promising intervention of MIA-induced psychiatric disorders in the offsprings.
Collapse
|
21
|
Bridi A, Bertolin K, Rissi VB, Mujica LKS, Glanzner WG, de Macedo MP, Comim FV, Gonçalves PBD, Antoniazzi AQ. Parthenogenetic bovine embryos secrete type I interferon capable of stimulating ISG15 in luteal cell culture. Anim Reprod 2018; 15:1268-1277. [PMID: 34221141 PMCID: PMC8203113 DOI: 10.21451/1984-3143-ar2018-0095] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Interferon tau (IFNT) is the pregnancy recognition signal in ruminants and is secreted by
trophoblast cells. Paracrine action in the endometrium is well established by inhibiting
luteolytic pulses of prostaglandin F2 alpha. Recently, endocrine action was documented
in the corpus luteum, blood cell and liver. It was hypothesized that conditioned medium (CM)
obtained from days 7, 9 and 12 parthenogenetic embryos alters luteal cell gene expression.
The aim was to establish a bovine mixed luteal cell culture to evaluate cellular response associated
to interferon stimulated genes, steroidogenesis and apoptosis. Conditioned medium was
obtained from Days 7, 9 and 12 parthenogenetic (PA) embryos culture. Moreover, antiviral
assay was performed on CM from Days 7, 9 and 12 to verify Type I interferon activity. Luteal cell
culture was validated by steroidogenic and apoptotic genes (CYP11A1
, HSD3B1, BAX, BCL2, AKT and
XIAP mRNA expression), and concentration of progesterone as endpoint. Luteal
cell culture was treated with interferon alpha (IFNA) and CM from parthenogenetic embryos.
Antiviral assay revealed Type I interferon activity on CM from embryos increasing on Days
9 and 12. ISG15 mRNA was greater in the mixed luteal cells culture treated
with 1, 10 and 100ng/ml of interferon alpha (IFNA) and also on Days 7, 9 and 12 CM treatments.
Concentration of progesterone was not altered in luteal cell culture regardless of treatments.
Steroidogenic and apoptotic genes were similar among groups in luteal cell culture treated
with different doses of IFNA or CM from PA embryos. In conclusion, parthenogenetic embryo-derived
CM has antiviral activity, luteal cell culture respond to Type I interferon by expressing
IGS15. These data indicate this model can be used for IFNT endocrine signaling studies.
Collapse
Affiliation(s)
- Alessandra Bridi
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Department of Large Animal Clinical Science, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Kalyne Bertolin
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Department of Large Animal Clinical Science, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vitor B Rissi
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Department of Large Animal Clinical Science, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Lady K S Mujica
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Department of Large Animal Clinical Science, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Werner G Glanzner
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Department of Large Animal Clinical Science, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Mariana P de Macedo
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Department of Large Animal Clinical Science, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Fabio V Comim
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Department of Large Animal Clinical Science, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Paulo B D Gonçalves
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Department of Large Animal Clinical Science, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Alfredo Q Antoniazzi
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Department of Large Animal Clinical Science, Federal University of Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
22
|
Zuo C, Sheng X, Ma M, Xia M, Ouyang L. ISG15 in the tumorigenesis and treatment of cancer: An emerging role in malignancies of the digestive system. Oncotarget 2018; 7:74393-74409. [PMID: 27626310 PMCID: PMC5342061 DOI: 10.18632/oncotarget.11911] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/01/2016] [Indexed: 02/07/2023] Open
Abstract
The interferon-stimulated gene 15 ubiquitin-like modifier (ISG15) encodes an IFN-inducible, ubiquitin-like protein. The ISG15 protein forms conjugates with numerous cellular proteins that are involved in a multitude of cellular functions, including interferon-induced immune responses and the regulation of cellular protein turnover. The expression of ISG15 and ISG15-mediated conjugation has been implicated in a wide range of human tumors and cancer cell lines, but the roles of ISG15 in tumorigenesis and responses to anticancer treatments remain largely unknown. In this review, we discuss the findings of recent studies with regard to the role of ISG15 pathways in cancers of the digestive system.
Collapse
Affiliation(s)
- Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Translation Medicine Research Center of Liver Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Graduate School, University of South China, Hengyang, Hunan, China
| | - Xinyi Sheng
- Graduate School, University of South China, Hengyang, Hunan, China
| | - Min Ma
- Department of Gastroduodenal and Pancreatic Surgery, Translation Medicine Research Center of Liver Cancer, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Man Xia
- Laboratory of Digestive Oncology, Hunan Province Cancer Institute, Changsha, Hunan, China
| | - Linda Ouyang
- Laboratory of Digestive Oncology, Hunan Province Cancer Institute, Changsha, Hunan, China
| |
Collapse
|
23
|
Desai S, Juncker M, Kim C. Regulation of mitophagy by the ubiquitin pathway in neurodegenerative diseases. Exp Biol Med (Maywood) 2018; 243:554-562. [PMID: 29316798 DOI: 10.1177/1535370217752351] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mitophagy is a cellular process by which dysfunctional mitochondria are degraded via autophagy. Increasing empirical evidence proposes that this mitochondrial quality-control mechanism is defective in neurons of patients with various neurodegenerative diseases such as Ataxia Telangiectasia, Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis. Accumulation of defective mitochondria and the production of reactive oxygen species due to defective mitophagy have been identified as causes underlying neurodegenerative disease pathogenesis. However, the reason mitophagy is defective in most neurodegenerative diseases is unclear. Like mitophagy, defects in the ubiquitin/26S proteasome pathway have been linked to neurodegeneration, resulting in the characteristic protein aggregates often seen in neurons of affected patients. Although initiation of mitophagy requires a functional ubiquitin pathway, whether defects in the ubiquitin pathway are causally responsible for defective mitophagy is not known. In this mini-review, we introduce mitophagy and ubiquitin pathways and provide a summary of our current understanding of the regulation of mitophagy by the ubiquitin pathway. We will then briefly review empirical evidence supporting mitophagy defects in neurodegenerative diseases. The review will conclude with a discussion of the constitutively elevated expression of ubiquitin-like protein Interferon-Stimulated Gene 15 (ISG15), an antagonist of the ubiquitin pathway, as a potential cause of defective mitophagy in neurodegenerative diseases. Impact statement Neurodegenerative diseases place an enormous burden on patients and caregivers globally. Over six million people in the United States alone suffer from neurodegenerative diseases, all of which are chronic, incurable, and with causes unknown. Identifying a common molecular mechanism underpinning neurodegenerative disease pathology is urgently needed to aid in the design of effective therapies to ease suffering, reduce economic cost, and improve the quality of life for these patients. Although the development of neurodegeneration may vary between neurodegenerative diseases, they have common cellular hallmarks, including defects in the ubiquitin-proteasome system and mitophagy. In this review, we will provide a summary of our current understanding of the regulation of mitophagy by the ubiquitin pathway and discuss the potential of targeting mitophagy and ubiquitin pathways for the treatment of neurodegeneration.
Collapse
Affiliation(s)
- Shyamal Desai
- Department of Biochemistry and Molecular Biology, LSUHSC-School of Medicine, New Orleans, LA 70112, USA
| | - Meredith Juncker
- Department of Biochemistry and Molecular Biology, LSUHSC-School of Medicine, New Orleans, LA 70112, USA
| | - Catherine Kim
- Department of Biochemistry and Molecular Biology, LSUHSC-School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
24
|
Dos Santos PF, Van Weyenbergh J, Delgobo M, Oliveira Patricio DD, Ferguson BJ, Guabiraba R, Dierckx T, Menezes SM, Báfica A, Mansur DS. ISG15-Induced IL-10 Is a Novel Anti-Inflammatory Myeloid Axis Disrupted during Active Tuberculosis. THE JOURNAL OF IMMUNOLOGY 2018; 200:1434-1442. [PMID: 29311364 DOI: 10.4049/jimmunol.1701120] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022]
Abstract
IFN-stimulated gene 15 (ISG15) deficiency in humans leads to severe IFNopathies and mycobacterial disease, the latter being previously attributed to its extracellular cytokine-like activity. In this study, we demonstrate a novel role for secreted ISG15 as an IL-10 inducer, unique to primary human monocytes. A balanced ISG15-induced monocyte/IL-10 versus lymphoid/IFN-γ expression, correlating with p38 MAPK and PI3K signaling, was found using targeted in vitro and ex vivo systems analysis of human transcriptomic datasets. The specificity and MAPK/PI3K-dependence of ISG15-induced monocyte IL-10 production was confirmed in vitro using CRISPR/Cas9 knockout and pharmacological inhibitors. Moreover, this ISG15/IL-10 axis was amplified in leprosy but disrupted in human active tuberculosis (TB) patients. Importantly, ISG15 strongly correlated with inflammation and disease severity during active TB, suggesting its potential use as a biomarker, awaiting clinical validation. In conclusion, this study identifies a novel anti-inflammatory ISG15/IL-10 myeloid axis that is disrupted in active TB.
Collapse
Affiliation(s)
- Paula Fernandes Dos Santos
- Laboratório de Imunobiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Santa Catarina CEP 88040-900, Brazil
| | - Johan Van Weyenbergh
- Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological Virology, KU Leuven - University of Leuven, 3000 Leuven, Belgium
| | - Murilo Delgobo
- Laboratório de Imunobiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Santa Catarina CEP 88040-900, Brazil
| | - Daniel de Oliveira Patricio
- Laboratório de Imunobiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Santa Catarina CEP 88040-900, Brazil
| | - Brian J Ferguson
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom; and
| | - Rodrigo Guabiraba
- Infectiologie et Santé Publique, Institut National de la Recherche Agronomique, Université François Rabelais de Tours, 37380 Nouzilly, France
| | - Tim Dierckx
- Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological Virology, KU Leuven - University of Leuven, 3000 Leuven, Belgium
| | - Soraya Maria Menezes
- Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological Virology, KU Leuven - University of Leuven, 3000 Leuven, Belgium
| | - André Báfica
- Laboratório de Imunobiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Santa Catarina CEP 88040-900, Brazil;
| | - Daniel Santos Mansur
- Laboratório de Imunobiologia, Departamento de Microbiologia, Imunologia e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Santa Catarina CEP 88040-900, Brazil;
| |
Collapse
|
25
|
Zhao P, Jiang T, Zhong Z, Zhao L, Yang S, Xia X. Inhibition of rabies virus replication by interferon-stimulated gene 15 and its activating enzyme UBA7. INFECTION GENETICS AND EVOLUTION 2017; 56:44-53. [PMID: 29056542 DOI: 10.1016/j.meegid.2017.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 10/19/2017] [Indexed: 01/27/2023]
Abstract
It was reported that ISG15 and its activating enzyme UBA7 have antiviral functions. However, there is no study to demonstrate whether ISG15 and UBA7 have anti-rabies virus function. In the current study, In vivo and in vitro anti-rabies virus function of ISG15 and UBA7 were investigated using RNAi technology. The results showed that shRNA knock-down of expression of ISG15 and UBA7 increased the titers of RABV in neuroblastoma cell line NA and microglial cell line BV-2 cells and shRNA knockdown of ISG15 conjugation alleviates the IFN-induced inhibition of RABV gene expression in vitro. Lentiviral vector mediated-shRNA knock-down of expression of ISG15 and UBA7 increased the titers of RABV in mouse brains and decreased the survivorship of mice. The study showed that ISG15 and UBA7 inhibit RABV replication in vitro and in vivo. To our knowledge, we for the first time documented the anti-RABV function of ISG15 and UBA7, which may provide a means of understanding the pathogenesis of rabies and improving therapeutic methods.
Collapse
Affiliation(s)
- Pingsen Zhao
- Clinical Core Laboratory, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou 514031, PR China; Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou 514031, PR China.
| | - Tianqi Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhixiong Zhong
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou 514031, PR China
| | - Lili Zhao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130122, China
| | - Songtao Yang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130122, China
| | - Xianzhu Xia
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun 130122, China
| |
Collapse
|
26
|
Kim CD, Reed RE, Juncker MA, Fang Z, Desai SD. Evidence for the Deregulation of Protein Turnover Pathways in Atm-Deficient Mouse Cerebellum: An Organotypic Study. J Neuropathol Exp Neurol 2017; 76:578-584. [DOI: 10.1093/jnen/nlx038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
27
|
Honke N, Shaabani N, Zhang DE, Hardt C, Lang KS. Multiple functions of USP18. Cell Death Dis 2016; 7:e2444. [PMID: 27809302 PMCID: PMC5260889 DOI: 10.1038/cddis.2016.326] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/12/2016] [Accepted: 09/16/2016] [Indexed: 12/12/2022]
Abstract
Since the discovery of the ubiquitin system and the description of its important role in the degradation of proteins, many studies have shown the importance of ubiquitin-specific peptidases (USPs). One special member of this family is the USP18 protein (formerly UBP43). In the past two decades, several functions of USP18 have been discovered: this protein is not only an isopeptidase but also a potent inhibitor of interferon signaling. Therefore, USP18 functions as 'a' maestro of many biological pathways in various cell types. This review outlines multiple functions of USP18 in the regulation of various immunological processes, including pathogen control, cancer development, and autoimmune diseases.
Collapse
Affiliation(s)
- Nadine Honke
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Namir Shaabani
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany.,Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dong-Er Zhang
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Cornelia Hardt
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Karl S Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| |
Collapse
|
28
|
Burks J, Reed RE, Desai SD. Free ISG15 triggers an antitumor immune response against breast cancer: a new perspective. Oncotarget 2016; 6:7221-31. [PMID: 25749047 PMCID: PMC4466680 DOI: 10.18632/oncotarget.3372] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/13/2015] [Indexed: 12/20/2022] Open
Abstract
Interferon-Stimulated Gene 15 (ISG15), an antagonist of the canonical ubiquitin pathway, is frequently overexpressed in various cancers. In cancer cells, ISG15 is detected as free (intracellular) and conjugated to cellular proteins (ISGylation). Free ISG15 is also secreted into the extracellular milieu. ISGylation has protumor functions and extracellular free ISG15 has immunomodulatory properties in vitro. Therefore, whether ISG15 is a tumor suppressor or tumor promoter in vivo remains controversial. The current study aimed to clarify the role of free ISG15 in tumorigenesis. Breast cancer cells stably expressing control, ISG15, and UbcH8 (ISG15-specific E2 ligase) shRNAs were used to assess the immunoregulatory and antitumor function of free ISG15 in cell culture (in vitro) and in nude mice (in vivo). We show that extracellular free ISG15 suppresses breast tumor growth and increases NK cell infiltration into xenografted breast tumors in nude mice, and intracellular free ISG15 enhances major histocompatibility complex (MHC) class I surface expression in breast cancer cells. We conclude that free ISG15 may have antitumor and immunoregulatory function in vivo. These findings provides the basis for developing strategies to increase systemic levels of free ISG15 to treat cancer patients overexpressing the ISG15 pathway.
Collapse
Affiliation(s)
- Julian Burks
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, New Orleans, LA, USA.,Present Address: Georgetown University Medical Center, Lombardi Comprehensive Cancer Center Department of Molecular Oncology, Washington, DC, USA
| | - Ryan E Reed
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, New Orleans, LA, USA
| | - Shyamal D Desai
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center-School of Medicine, New Orleans, LA, USA
| |
Collapse
|
29
|
Romero JJ, Antoniazzi AQ, Nett TM, Ashley RL, Webb BT, Smirnova NP, Bott RC, Bruemmer JE, Bazer FW, Anthony RV, Hansen TR. Temporal Release, Paracrine and Endocrine Actions of Ovine Conceptus-Derived Interferon-Tau During Early Pregnancy. Biol Reprod 2015; 93:146. [PMID: 26559679 DOI: 10.1095/biolreprod.115.132860] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/03/2015] [Indexed: 11/01/2022] Open
Abstract
The antiviral activity of interferon (IFN) increases in uterine vein serum (UVS) during early pregnancy in sheep. This antiviral activity in UVS collected on Day 15 of pregnancy is blocked by anti-IFN-tau (anti-IFNT) antibodies. Conceptus-derived IFNT was hypothesized to induce IFN-stimulated gene (ISG) expression in endometrium and extrauterine tissues during pregnancy. To test this hypothesis, blood was collected from ewes on Days 12-16 of the estrous cycle or pregnancy. Serum progesterone was >1.7 ng/ml in pregnant (P) and nonpregnant (NP) ewes until Day 13, then declined to <0.6 ng/ml by Day 15 in NP ewes. A validated IFNT radioimmunoassay detected IFNT in uterine flushings (UFs) on Days 13-16 and in UVS on Days 15-16 of pregnancy. IFNT detection in UF correlated with paracrine induction of ISGs in the endometrium and occurred prior to the inhibition of estrogen receptor 1 and oxytocin receptor expression in uterine epithelia on Day 14 of pregnancy. Induction of ISG mRNAs in corpus luteum (CL) and liver tissue occurred by Day 14 and in peripheral blood mononuclear cells by Day 15 in P ewes. Expression of mRNAs for IFN signal transducers and ISGs were greater in the CL of P than that of NP ewes on Day 14. It is concluded that: 1) paracrine actions of IFNT coincide with detection of IFNT in UF; 2) endocrine action of IFNT ensues through induction of ISGs in peripheral tissues; and 3) IFNT can be detected in UVS, but not until Days 15-16 of pregnancy, which may be limited by the sensitivity of the IFNT radioimmunoassay.
Collapse
Affiliation(s)
- Jared J Romero
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Alfredo Q Antoniazzi
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Terry M Nett
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Ryan L Ashley
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Brett T Webb
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Natalia P Smirnova
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Rebecca C Bott
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Jason E Bruemmer
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Fuller W Bazer
- Department of Animal Sciences, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Russell V Anthony
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Thomas R Hansen
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
30
|
Kaposi's Sarcoma-Associated Herpesvirus Viral Interferon Regulatory Factor 1 Interacts with a Member of the Interferon-Stimulated Gene 15 Pathway. J Virol 2015; 89:11572-83. [PMID: 26355087 DOI: 10.1128/jvi.01482-15] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/01/2015] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED Kaposi's sarcoma-associated herpesvirus (KSHV) is a gammaherpesvirus known to establish lifelong latency in the human host. We and others have previously shown that three KSHV homologs of cellular interferon regulatory factors (IRFs), known as viral IRFs (vIRFs), participate in evasion of the host interferon (IFN) response. We report that vIRF1 interacts with the cellular interferon-stimulated gene 15 (ISG15) E3 ligase, HERC5, in the context of Toll-like receptor 3 (TLR3) activation and IFN induction. The ISG15 protein is covalently conjugated to target proteins upon activation of the interferon response. Interaction between vIRF1 and HERC5 was confirmed by immunoprecipitation, and the region between amino acids 224 and 349 of vIRF1 was required for interaction with HERC5. We further report that expression of vIRF1 in the context of TLR3 activation results in decreased ISG15 conjugation of proteins. Specifically, TLR3-induced ISG15 conjugation and protein levels of cellular IRF3, a known ISG15 target, were decreased in the presence of vIRF1 compared to the control. vIRF1 itself was also identified as a target of ISG15 conjugation. KSHV-infected cells exhibited increased ISG15 conjugation upon reactivation from latency in coordination with increased IFN. Furthermore, knockdown of ISG15 in latently infected cells resulted in a higher level of KSHV reactivation and an increase in infectious virus. These data suggest that the KSHV vIRF1 protein affects ISG15 conjugation and interferon responses and may contribute to effective KSHV replication. IMPORTANCE The KSHV vIRF1 protein can inhibit interferon activation in response to viral infection. We identified a cellular protein named HERC5, which is the major ligase for ISG15, as a vIRF1 binding partner. vIRF1 association with HERC5 altered ISG15 modification of cellular proteins, and knockdown of ISG15 augmented reactivation of KSHV from latency.
Collapse
|
31
|
Schmaltz-Panneau B, Cordova A, Dhorne-Pollet S, Hennequet-Antier C, Uzbekova S, Martinot E, Doret S, Martin P, Mermillod P, Locatelli Y. Early bovine embryos regulate oviduct epithelial cell gene expression during in vitro co-culture. Anim Reprod Sci 2014; 149:103-16. [DOI: 10.1016/j.anireprosci.2014.06.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 05/26/2014] [Accepted: 06/19/2014] [Indexed: 01/12/2023]
|
32
|
Hansen TR, Pru JK. ISGylation: a conserved pathway in mammalian pregnancy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 759:13-31. [PMID: 25030758 DOI: 10.1007/978-1-4939-0817-2_2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Successful pregnancy includes remodeling and differentiation of the endometrium in response to sex steroid hormones, development of maternal immunotolerance to the implanting embryo, and modification of the local uterine environment by the embryo to suit its own needs. The major signal released by the ruminant conceptus during establishment of pregnancy is interferon-tau (IFNT) that stimulates the expression of many genes in the endometrium and ovary. One of these genes is called interferon stimulated gene 15 (ISG15), which encodes a ubiquitin homolog with a C-terminal Gly that becomes covalently attached to Lys residues on targeted proteins through an ATP-dependent multi-step enzymatic reaction called ISGylation. The conceptus-derived induction of endometrial ISGs also occurs in mouse and human deciduas and placenta, in response to pregnancy presumably through action of cytokines such as interleukins and type I IFN. Described herein is evidence to support the concept that ISGylation is a maternal response to the developing conceptus, implantation and placentation that is conserved across mammalian pregnancy. Although the precise role for ISG15 remains elusive during pregnancy, it is clear that up-regulation in response to pregnancy may impart a pre-emptive defense to infection or other environmental insults, and protection of the conceptus against inflammatory insults across species.
Collapse
Affiliation(s)
- Thomas R Hansen
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3197 Rampart Road, Fort Collins, CO, 80521, USA,
| | | |
Collapse
|
33
|
Zhao H, Yang J, Li K, Ding X, Lin R, Ma Y, Liu J, Zhong Z, Qian X, Bo X, Zhou Z, Wang S. Proteomic analysis at the subcellular level for host targets against influenza A virus (H1N1). Antiviral Res 2013; 100:673-87. [PMID: 24161511 DOI: 10.1016/j.antiviral.2013.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 10/10/2013] [Accepted: 10/14/2013] [Indexed: 01/28/2023]
Abstract
Influenza viruses (IVs) trigger a series of intracellular signaling events and induce complex cellular responses from the infected host cell. Accumulating evidence suggests that host cell proteins play an essential role in viral propagation and represent novel antiviral therapeutic targets. Subcellular proteomic technology provides a method for understanding regional differences at the protein level. The present study, which utilized subcellular proteomic technology, aimed to identify host cell proteins involved in influenza virus (HIN1) infection. Two-dimensional gel electrophoresis (2-DE) combined with mass spectrum (MS) was performed on protein extracts from the nuclei, cytoplasm, and mitochondria of infected and control human lung epithelial cells (A549). In total, 112 differentially expressed protein molecules were identified; 80 protein spots were successfully validated using MS. The differential expression of ISG15, MIF, PDCD5, and UCHL1 was confirmed by western blot. Furthermore, antisense oligodeoxyribonucleotide (ODN) targeting ISG15, MIF, PDCD5, and UCHL1 significantly mitigated HIN1 propagation, cytopathic effects, vRNA by RT-qPCR, and rescued cell viability in A549 cells. Taken together, the differentially expressed proteins identified in this study might provide novel targets for anti-influenza drug development.
Collapse
Affiliation(s)
- Haibao Zhao
- Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
microRNA control of interferons and interferon induced anti-viral activity. Mol Immunol 2013; 56:781-93. [PMID: 23962477 DOI: 10.1016/j.molimm.2013.07.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 07/11/2013] [Accepted: 07/14/2013] [Indexed: 12/22/2022]
Abstract
Interferons (IFNs) are cytokines that are spontaneously produced in response to virus infection. They act by binding to IFN-receptors (IFN-R), which trigger JAK/STAT cell signalling and the subsequent induction of hundreds of IFN-inducible genes, including both protein-coding and microRNA genes. IFN-induced genes then act synergistically to prevent virus replication and create an anti-viral state. miRNA are therefore integral to the innate response to virus infection and are important components of IFN-mediated biology. On the other hand viruses also encode miRNAs that in some cases interfere directly with the IFN response to infection. This review summarizes the important roles of miRNAs in virus infection acting both as IFN-stimulated anti-viral molecules and as critical regulators of IFNs and IFN-stimulated genes. It also highlights how recent knowledge in RNA editing influence miRNA control of virus infection.
Collapse
|
35
|
Zebrafish ISG15 exerts a strong antiviral activity against RNA and DNA viruses and regulates the interferon response. J Virol 2013; 87:10025-36. [PMID: 23824820 DOI: 10.1128/jvi.01294-12] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
ISG15, a 15-kDa interferon-induced protein that participates in antiviral defenses of mammals, is highly conserved among vertebrates. In fish, as in mammals, viral infection and interferon treatment induce isg15 expression. The two ubiquitin-like domains of ISG15 and the presence of a consensus LRLRGG sequence in the C-terminal region, which is required for the covalent conjugation to a substrate protein, are also conserved in fish. Our data demonstrate that overexpression of zebrafish ISG15 (zf-ISG15) in EPC cells is sufficient to inhibit viral infection by RNA viruses belonging to the genera Novirhabdovirus and Birnavirus and by DNA viruses of the genus Iridovirus. In coexpression experiments with IHNV proteins, we demonstrate specific ISGylation of phosphoprotein and nonvirion protein. Mutation of the glycine residues in the consensus LRLRGG motif abolishes zf-ISG15 conjugation to these proteins and the cellular protection against viral infection, thus connecting ISGylation and ISG15-dependent viral restriction. Additionally, zf-ISG15 overexpression triggers induction of the rig-I and viperin genes as well as, to a lesser extent, the IFN gene. Overall, our data demonstrate the antiviral effect of a fish ISG15 protein, revealing the conservation among vertebrates of an ISGylation mechanism likely directed against viruses. Furthermore, our findings indicate that zf-ISG15 affects the IFN system at several levels, and its study shall shed further light on the evolution of the complex regulation of the innate antiviral response in vertebrate cells.
Collapse
|
36
|
Desai SD, Reed RE, Babu S, Lorio EA. ISG15 deregulates autophagy in genotoxin-treated ataxia telangiectasia cells. J Biol Chem 2012; 288:2388-402. [PMID: 23212917 DOI: 10.1074/jbc.m112.403832] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Ataxia-telangiectasia (A-T) is a cerebellar neurodegenerative disorder; however, the basis for the neurodegeneration in A-T is not well established. Lesions in the ubiquitin and autophagy pathways are speculated to contribute to the neurodegeneration in other neurological diseases and may have a role in A-T neurodegeneration. Our recent studies revealed that the constitutively elevated ISG15 pathway impairs targeted proteasome-mediated protein degradation in A-T cells. Here, we demonstrate that the basal autophagy pathway is activated in the ubiquitin pathway-compromised A-T cells. We also show that genotoxic stress triggers aberrant degradation of the proteasome and autophagy substrates (autophagic flux) in A-T cells. Inhibition of autophagy at an early stage using 3-methyladenine blocked UV-induced autophagic flux in A-T cells. On the other hand, bafilomycin A1, which inhibits autophagy at a late stage, failed to block UV-induced autophagic flux, suggesting that overinduction of autophagy may underlie aberrant autophagic flux in A-T cells. The ISG15-specific shRNA that restored proteasome function restores autophagic function in A-T cells. These findings suggest that autophagy compensates for the ISG15-dependent ablation of proteasome-mediated protein degradation in A-T cells. Genotoxic stress overactivates this compensatory mechanism, triggering aberrant autophagic flux in A-T cells. Supporting the model, we show that autophagy is activated in the brain tissues of human A-T patients. This highlights a plausible causal contribution of a novel "ISG15 proteinopathy" in A-T neuronal cell death.
Collapse
Affiliation(s)
- Shyamal D Desai
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center School of Medicine, New Orleans, Louisiana 70112, USA
| | | | | | | |
Collapse
|
37
|
Abstract
The highly conserved ubiquitin polypeptide functions by covalently modifying other proteins. This modification has a well-established role in facilitating substrate degradation by the proteasome and can regulate some proteins by ways other than targeting them to the proteasome. It has now emerged that proteins bearing only distant similarity to ubiquitin can also be attached to specific proteins. The consequences of most of these modifications are not yet understood. However, two recent papers on one ubiquitin-like protein, SUMO-1, demonstrate a role in targeting a protein crucial for nucleocytoplasmic trafficking to the nuclear pore complex. These and other recent findings suggest a much wider influence of the 'ubiquitin system' on cell biology and raise intriguing regulatory and mechanistic questions.
Collapse
|
38
|
Jain A, Baviskar PS, Kandasamy S, Kumar R, Singh R, Kumar S, Agarwal SK, Joshi P, Mitra A. Interferon stimulated gene 15 (ISG15): Molecular characterization and expression profile in endometrium of buffalo (Bubalus bubalis). Anim Reprod Sci 2012; 133:159-68. [DOI: 10.1016/j.anireprosci.2012.06.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 06/28/2012] [Accepted: 06/29/2012] [Indexed: 11/17/2022]
|
39
|
|
40
|
Interplay between interferon-mediated innate immunity and porcine reproductive and respiratory syndrome virus. Viruses 2012; 4:424-46. [PMID: 22590680 PMCID: PMC3347317 DOI: 10.3390/v4040424] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/15/2012] [Accepted: 03/18/2012] [Indexed: 12/24/2022] Open
Abstract
Innate immunity is the first line of defense against viral infection, and in turn, viruses have evolved to evade host immune surveillance. As a result, viruses may persist in host and develop chronic infections. Type I interferons (IFN-α/β) are among the most potent antiviral cytokines triggered by viral infections. Porcine reproductive and respiratory syndrome (PRRS) is a disease of pigs that is characterized by negligible induction of type I IFNs and viral persistence for an extended period. For IFN production, RIG-I/MDA5 and JAK-STAT pathways are two major signaling pathways, and recent studies indicate that PRRS virus is armed to modulate type I IFN responses during infection. This review describes the viral strategies for modulation of type I IFN responses. At least three non-structural proteins (Nsp1, Nsp2, and Nsp11) and a structural protein (N nucleocapsid protein) have been identified and characterized to play roles in the IFN suppression and NF-κB pathways. Nsp's are early proteins while N is a late protein, suggesting that additional signaling pathways may be involved in addition to the IFN pathway. The understanding of molecular bases for virus-mediated modulation of host innate immune signaling will help us design new generation vaccines and control PRRS.
Collapse
|
41
|
Desai SD, Reed RE, Burks J, Wood LM, Pullikuth AK, Haas AL, Liu LF, Breslin JW, Meiners S, Sankar S. ISG15 disrupts cytoskeletal architecture and promotes motility in human breast cancer cells. Exp Biol Med (Maywood) 2011; 237:38-49. [PMID: 22185919 DOI: 10.1258/ebm.2011.011236] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The interferon-stimulated gene 15 (ISG15) pathway is highly elevated in breast cancer; however, very little is known about how the ISG15 pathway contributes to breast tumorigenesis. In the current study, using the gene disruption approach, we demonstrate that both ISG15 and UbcH8 (ISG15-specific conjugating enzyme) disrupt F-actin architecture and formation of focal adhesions in ZR-75-1 breast cancer cells. In addition, ISG15 and UbcH8 promote breast cancer cell migration. We also demonstrate that ISG15 inhibits ubiquitin/26S proteasome-mediated turnover of proteins implicated in tumor cell motility, invasion and metastasis. Together, our results suggest that the aberrant activation of the ISG15 pathway confers a motile phenotype to breast cancer cells by disrupting cell architecture and stabilizing proteins involved in cell motility, invasion and metastasis. Because the cellular architecture is conserved and the ISG15 pathway is constitutively activated in tumor cells of different lineages, it is reasonable to assume that our observations in breast cancer must hold true for many other tumors.
Collapse
Affiliation(s)
- Shyamal D Desai
- Department of Biochemistry and Molecular Biology, LSU Health Sciences Center-School of Medicine, 1901 Perdido Street, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wood LM, Sankar S, Reed RE, Haas AL, Liu LF, McKinnon P, Desai SD. A novel role for ATM in regulating proteasome-mediated protein degradation through suppression of the ISG15 conjugation pathway. PLoS One 2011; 6:e16422. [PMID: 21298066 PMCID: PMC3027683 DOI: 10.1371/journal.pone.0016422] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 12/15/2010] [Indexed: 01/12/2023] Open
Abstract
Ataxia Telangiectasia (A-T) is an inherited immunodeficiency disorder wherein mutation of the ATM kinase is responsible for the A-T pathogenesis. Although the precise role of ATM in A-T pathogenesis is still unclear, its function in responding to DNA damage has been well established. Here we demonstrate that in addition to its role in DNA repair, ATM also regulates proteasome-mediated protein turnover through suppression of the ISG15 pathway. This conclusion is based on three major pieces of evidence: First, we demonstrate that proteasome-mediated protein degradation is impaired in A-T cells. Second, we show that the reduced protein turnover is causally linked to the elevated expression of the ubiquitin-like protein ISG15 in A-T cells. Third, we show that expression of the ISG15 is elevated in A-T cells derived from various A-T patients, as well as in brain tissues derived from the ATM knockout mice and A-T patients, suggesting that ATM negatively regulates the ISG15 pathway. Our current findings suggest for the first time that proteasome-mediated protein degradation is impaired in A-T cells due to elevated expression of the ISG15 conjugation pathway, which could contribute to progressive neurodegeneration in A-T patients.
Collapse
Affiliation(s)
- Laurence M. Wood
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Surendran Sankar
- Department of Biochemistry and Molecular Biology, Louisiana State University of Health Sciences Center-School of Medicine, New Orleans, Louisiana, United States of America
| | - Ryan E. Reed
- Department of Biochemistry and Molecular Biology, Louisiana State University of Health Sciences Center-School of Medicine, New Orleans, Louisiana, United States of America
| | - Arthur L. Haas
- Department of Biochemistry and Molecular Biology, Louisiana State University of Health Sciences Center-School of Medicine, New Orleans, Louisiana, United States of America
| | - Leroy F. Liu
- Department of Pharmacology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey, United States of America
| | - Peter McKinnon
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Shyamal D. Desai
- Department of Biochemistry and Molecular Biology, Louisiana State University of Health Sciences Center-School of Medicine, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
43
|
Antiviral Properties of ISG15. Viruses 2010; 2:2154-2168. [PMID: 21994614 PMCID: PMC3185569 DOI: 10.3390/v2102154] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 09/08/2010] [Accepted: 09/15/2010] [Indexed: 12/04/2022] Open
Abstract
The type I interferon system plays a critical role in limiting the spread of viral infection. Viruses induce the production of interferon (IFN), which after binding to the IFN-α/β receptor (IFNAR), and triggering of the JAK/STAT signaling cascade, results in the induction of interferon-stimulated genes (ISGs). These ISGs function to inhibit viral replication and to regulate the host immune response. Among these ISGs, the ubiquitin-like molecule, ISG15, is one of the most strongly induced proteins. Similar to ubiquitin, through an IFN induced conjugation cascade, ISG15 is covalently linked to a variety of cellular proteins, suggesting regulation of different cellular processes. Studies performed over the past several years have shown that ISG15 plays a central role in the host’s antiviral response against many viruses. Mice lacking ISG15 display increased susceptibility to multiple viruses. Furthermore, several viruses have developed immune evasion strategies that directly target the ISG15 pathway. Work is now underway to determine the mechanism by which ISG15 functions as an antiviral molecule, such that therapies targeting this pathway can be developed in the future.
Collapse
|
44
|
Ashley RL, Henkes LE, Bouma GJ, Pru JK, Hansen TR. Deletion of the Isg15 gene results in up-regulation of decidual cell survival genes and down-regulation of adhesion genes: implication for regulation by IL-1beta. Endocrinology 2010; 151:4527-36. [PMID: 20660068 DOI: 10.1210/en.2010-0166] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ubiquitin homolog interferon stimulated gene 15 (ISG15) is up-regulated in the endometrium in response to pregnancy in primates, ruminants, pigs, and mice. ISG15 covalently attaches to intracellular proteins (isgylation) and regulates numerous intracellular responses. We hypothesized that ISG15 depletion (Isg15(-/-)) alters decidual tissue gene expression and that IL-1beta induces ISG15 expression and isgylation in cultured murine decidual explants and human uterine fibroblasts (HuFs). After studying the reproductive phenotype, contrary to earlier reports, up to 50% of the fetuses die between 7.5 and 12.5 d post coitum (dpc) in Isg15(-/-) mothers when mated to Isg15(-/-) fathers. Using microarray analysis, over 500 genes are differentially regulated in 7.5 dpc deciduas from Isg15(-/-) compared with Isg15(+/+) mice. The gene for interferon-inducible protein 202b, which functions in cell-survival mechanisms, was up-regulated (mRNA and protein) in deciduas from Isg15(-/-) mice. Culture of Isg15(+/+) mouse decidual explants (7.5 dpc) with IL-1beta decreased Isg15 mRNA but increased free and conjugated ISG15. In predecidual HuF cells, IL-1beta treatment increased ISG15 mRNA and isgylation. Additionally, IL-1beta up-regulated expression of enzymes (HERC5, UBCH8) that coordinate the covalent conjugation of ISG15 to target proteins, as well as the gene that encodes the deisglyation enzyme UBP43 in HuF cells. In conclusion, deletion of Isg15 gene results in 50% fetal loss after 7.5 dpc, which can be explained through differential decidual gene expression that is functionally tied to cell survival and adhesion pathways. This fetal death also might relate to impaired IL-1beta signaling, because ISG15 and isgylation are induced by IL-1beta in human and murine endometrial stromal cells.
Collapse
Affiliation(s)
- Ryan L Ashley
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | | | | | | | | |
Collapse
|
45
|
Heim MH. HCV innate immune responses. Viruses 2009; 1:1073-88. [PMID: 21994583 PMCID: PMC3185522 DOI: 10.3390/v1031073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 11/25/2009] [Accepted: 11/26/2009] [Indexed: 01/07/2023] Open
Abstract
Hepatitis C virus (HCV) establishes a persistent infection in more than 70% of infected individuals. This striking ability to evade the powerful innate immune system results from viral interference occurring at several levels of the interferon (IFN) system. There is strong evidence from cell culture experiments that HCV can inhibit the induction of IFNβ by cleaving important proteins in the virus sensory pathways of cells such as MAVS and TRIF. There is also evidence that HCV interferes with IFNα signaling through the Jak-STAT pathway, and that HCV proteins target IFN effector systems such as protein kinase R (PKR). These in vitro findings will have to be confirmed in clinical trials investigating the molecular mechanisms of HCV interference with the innate immune system in liver samples.
Collapse
Affiliation(s)
- Markus H. Heim
- Clinic for Gastroenterology and Hepatology, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
- Department of Biomedicine, University Basel, ZLF, Hebelstrasse 20, CH-4031 Basel, Switzerland; E-Mail: ; Tel.: +41 61 265 25 25; Fax: +41-61-265 52 53
| |
Collapse
|
46
|
Interferon-induced ISG15 conjugation inhibits influenza A virus gene expression and replication in human cells. J Virol 2009; 83:5971-7. [PMID: 19357168 DOI: 10.1128/jvi.01667-08] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ubiquitin-like ISG15 protein, as well as its conjugating enzymes, is induced by type I interferons (IFNs). Experiments using ISG15 knockout (ISG15(-/-)) mice established that ISG15 and/or its conjugation inhibits the replication of influenza A virus. However, in contrast to the virus inhibition results for mice, the rates of virus replication in ISG15(+/+) and ISG15(-/-) mouse embryo fibroblasts in tissue culture were similar. Here we focus on human tissue culture cells and on the effect of ISG15 and/or its conjugation on influenza A virus gene expression and replication in such cells. We demonstrate that IFN-induced antiviral activity against influenza A virus in human cells is significantly alleviated by inhibiting ISG15 conjugation using small interfering RNAs directed against ISG15-conjugating enzymes. IFN-induced antiviral activity against influenza A virus protein synthesis was reduced 5- to 20-fold by suppressing ISG15 conjugation. The amounts of the viral proteins that were restored by these siRNA treatments were approximately 40 to 50% of the amounts produced in cells that were not pretreated with IFN. Further, we show that ISG15 conjugation inhibits influenza A virus replication 10- to 20-fold at early times after infection in human cells. These results show that ISG15 conjugation plays a substantial role in the antiviral state induced by IFN in human cells. In contrast, we show that in mouse embryo fibroblasts ISG15 conjugation not only does not affect influenza A virus replication but also does not contribute to the IFN-induced antiviral activity against influenza A virus gene expression.
Collapse
|
47
|
Shoemaker ML, Smirnova NP, Bielefeldt-Ohmann H, Austin KJ, van Olphen A, Clapper JA, Hansen TR. Differential expression of the type I interferon pathway during persistent and transient bovine viral diarrhea virus infection. J Interferon Cytokine Res 2009; 29:23-35. [PMID: 19014339 DOI: 10.1089/jir.2008.0033] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Persistent infection with bovine viral diarrhea virus (BVDV) serves as a reservoir for the perpetuation of infection in cattle populations and causes a range of adverse effects on the health of the host. To study the interactions of the virus with the host, gene expression was compared in the blood of persistently infected (PI) and uninfected steer, and in the blood and tissues of PI fetuses, transiently infected (TI), and uninfected bovine fetuses. Microarray analysis of PI steer blood revealed differential gene expression indicative of an interferon (IFN) response including genes involved in cell cycle regulation, which may contribute to long-term adverse effects. Upregulation of IFN-stimulated genes (e.g., ISG15, PKR) and RNA helicases (RIG-I, LGP2, MDA-5) was identified in both PI fetal and steer blood in comparison to controls, indicating a continued stimulation of the innate antiviral response as a result of the persistent viremia. ISG15 was studied in further detail, implicating reticular cells as basal producers of ISG15 in the spleen, in addition to endothelial and macrophage-like cells in infected spleen. Consequences of chronic IFN pathway activation in PI cattle may include growth- and immunosuppression, the pathogenesis of which is still poorly understood. This study lends new insights into the interactions between BVDV and its host, and can serve as a model for studies of the role of the IFN system in persistent infections.
Collapse
Affiliation(s)
- Megan L Shoemaker
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, Fort Collins, Colorado 80523, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Giannakopoulos NV, Arutyunova E, Lai C, Lenschow DJ, Haas AL, Virgin HW. ISG15 Arg151 and the ISG15-conjugating enzyme UbE1L are important for innate immune control of Sindbis virus. J Virol 2009; 83:1602-10. [PMID: 19073728 PMCID: PMC2643764 DOI: 10.1128/jvi.01590-08] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Accepted: 12/02/2008] [Indexed: 01/14/2023] Open
Abstract
Interferon (IFN)-stimulated gene 15 (ISG15) is a ubiquitin-like molecule that conjugates to target proteins via a C-terminal LRLRGG motif and has antiviral function in vivo. We used structural modeling to predict human ISG15 (hISG15) residues important for interacting with its E1 enzyme, UbE1L. Kinetic analysis revealed that mutation of arginine 153 to alanine (R153A) ablated hISG15-hUbE1L binding and transthiolation of UbcH8. Mutation of other predicted UbE1L-interacting residues had minimal effects on the transfer of ISG15 from UbE1L to UbcH8. The capacity of hISG15 R153A to form protein conjugates in 293T cells was markedly diminished. Mutation of the homologous residue in mouse ISG15 (mISG15), arginine 151, to alanine (R151A) also attenuated protein ISGylation following transfection into 293T cells. We assessed the role of ISG15-UbE1L interactions in control of virus infection by constructing double subgenomic Sindbis viruses that expressed the mISG15 R151A mutant. While expression of mISG15 protected alpha/beta-IFN-receptor-deficient (IFN-alphabetaR(-/-)) mice from lethality following Sindbis virus infection, expression of mISG15 R151A conferred no survival benefit. The R151A mutation also attenuated ISG15's ability to decrease Sindbis virus replication in IFN-alphabetaR(-/-) mice or prolong survival of ISG15(-/-) mice. The importance of UbE1L was confirmed by demonstrating that mice lacking this ISG15 E1 enzyme were highly susceptible to Sindbis virus infection. Together, these data support a role for protein conjugation in the antiviral effects of ISG15.
Collapse
Affiliation(s)
- Nadia V Giannakopoulos
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
49
|
Kim JH, Luo JK, Zhang DE. The Level of Hepatitis B Virus Replication Is Not Affected by Protein ISG15 Modification but Is Reduced by Inhibition of UBP43 (USP18) Expression. THE JOURNAL OF IMMUNOLOGY 2008; 181:6467-72. [DOI: 10.4049/jimmunol.181.9.6467] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
50
|
Guerra S, Cáceres A, Knobeloch KP, Horak I, Esteban M. Vaccinia virus E3 protein prevents the antiviral action of ISG15. PLoS Pathog 2008; 4:e1000096. [PMID: 18604270 PMCID: PMC2434199 DOI: 10.1371/journal.ppat.1000096] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Accepted: 05/29/2008] [Indexed: 01/15/2023] Open
Abstract
The ubiquitin-like modifier ISG15 is one of the most predominant proteins induced by type I interferons (IFN). In this study, murine embryo fibroblast (MEFs) and mice lacking the gene were used to demonstrate a novel role of ISG15 as a host defense molecule against vaccinia virus (VACV) infection. In MEFs, the growth of replication competent Western Reserve (WR) VACV strain was affected by the absence of ISG15, but in addition, virus lacking E3 protein (VVDeltaE3L) that is unable to grow in ISG15+/+ cells replicated in ISG15-deficient cells. Inhibiting ISG15 with siRNA or promoting its expression in ISG15-/- cells with a lentivirus vector showed that VACV replication was controlled by ISG15. Immunoprecipitation analysis revealed that E3 binds ISG15 through its C-terminal domain. The VACV antiviral action of ISG15 and its interaction with E3 are events independent of PKR (double-stranded RNA-dependent protein kinase). In mice lacking ISG15, infection with VVDeltaE3L caused significant disease and mortality, an effect not observed in VVDeltaE3L-infected ISG15+/+ mice. Pathogenesis in ISG15-deficient mice infected with VVDeltaE3L or with an E3L deletion mutant virus lacking the C-terminal domain triggered an enhanced inflammatory response in the lungs compared with ISG15+/+-infected mice. These findings showed an anti-VACV function of ISG15, with the virus E3 protein suppressing the action of the ISG15 antiviral factor.
Collapse
Affiliation(s)
- Susana Guerra
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, Madrid, Spain
- Department of Preventive Medicine and Public Health, Universidad Autónoma, Madrid, Spain
- * E-mail: (SG); (ME)
| | - Ana Cáceres
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, Madrid, Spain
| | - Klaus-Peter Knobeloch
- Abteilung Molekulare Genetik, Leibniz Institut fur Molekulare Pharmakologie, Berlin, Germany
| | - Ivan Horak
- Abteilung Molekulare Genetik, Leibniz Institut fur Molekulare Pharmakologie, Berlin, Germany
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología CSIC, Campus Universidad Autónoma, Madrid, Spain
- * E-mail: (SG); (ME)
| |
Collapse
|