1
|
Mbani CJ, Morvan C, Nekoua MP, Debuysschere C, Alidjinou EK, Moukassa D, Hober D. Enterovirus Antibodies: Friends and Foes. Rev Med Virol 2024; 34:e70004. [PMID: 39505825 DOI: 10.1002/rmv.70004] [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: 07/10/2024] [Revised: 10/02/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024]
Abstract
Enteroviruses (EV) initiate replication by binding to their cellular receptors, leading to the uncoating and release of the viral genome into the cytosol of the host cell. Neutralising antibodies (NAbs) binding to epitopes on enteroviral capsid proteins can inhibit this infectious process through several mechanisms of neutralisation in vitro. Fc-mediated antibody effector functions such as antibody-dependent cell-mediated cytotoxicity and antibody-dependent cellular phagocytosis have also been described for some EV. However, antibody binding to virions does not always result in viral neutralisation. Non-neutralising antibodies, or sub-neutralising concentrations of antibodies, can enhance infection of viruses, leading to more severe pathologies. This phenomenon, known as antibody-dependent enhancement (ADE) of infection, has been described in vitro and/or in vivo for EV including poliovirus, coxsackievirus B and EV-A71. It has been shown that ADE of EV infection is mediated by FcγRs expressed by monocytes, macrophages, B lymphocytes and granulocytes. Antibodies play a crucial role in the diagnosis and monitoring of infections. They are valuable markers that have been used to establish a link between enteroviral infection and chronic diseases such as type 1 diabetes. Monoclonal and polyclonal antibodies targeting enteroviral proteins have been developed and shown to be effective to prevent or combat EV infections in vitro and in vivo. In addition, vaccines are under development, and clinical trials of vaccines are underway or have been completed, providing hope for the prevention of diseases due to EV. However, the ADE of the infection should be considered in the development of anti-EV antibodies or safe vaccines.
Collapse
Affiliation(s)
- Chaldam Jespère Mbani
- Laboratoire de Virologie URL3610, Univ. Lille et CHU Lille, Lille, France
- Laboratoire de Biologie Cellulaire et Moléculaire, Faculté des Sciences et Techniques, Université Marien Ngouabi, Brazzaville, Congo
| | - Corentin Morvan
- Laboratoire de Virologie URL3610, Univ. Lille et CHU Lille, Lille, France
| | | | - Cyril Debuysschere
- Laboratoire de Virologie URL3610, Univ. Lille et CHU Lille, Lille, France
| | | | - Donatien Moukassa
- Laboratoire de Biologie Cellulaire et Moléculaire, Faculté des Sciences et Techniques, Université Marien Ngouabi, Brazzaville, Congo
| | - Didier Hober
- Laboratoire de Virologie URL3610, Univ. Lille et CHU Lille, Lille, France
| |
Collapse
|
2
|
Garrison AR, Moresco V, Zeng X, Cline CR, Ward MD, Ricks KM, Olschner SP, Cazares LH, Karaaslan E, Fitzpatrick CJ, Bergeron É, Pegan SD, Golden JW. Nucleocapsid protein-specific monoclonal antibodies protect mice against Crimean-Congo hemorrhagic fever virus. Nat Commun 2024; 15:1722. [PMID: 38409240 PMCID: PMC10897337 DOI: 10.1038/s41467-024-46110-4] [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: 08/22/2023] [Accepted: 02/07/2024] [Indexed: 02/28/2024] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a WHO priority pathogen. Antibody-based medical countermeasures offer an important strategy to mitigate severe disease caused by CCHFV. Most efforts have focused on targeting the viral glycoproteins. However, glycoproteins are poorly conserved among viral strains. The CCHFV nucleocapsid protein (NP) is highly conserved between CCHFV strains. Here, we investigate the protective efficacy of a CCHFV monoclonal antibody targeting the NP. We find that an anti-NP monoclonal antibody (mAb-9D5) protected female mice against lethal CCHFV infection or resulted in a significant delay in mean time-to-death in mice that succumbed to disease compared to isotype control animals. Antibody protection is independent of Fc-receptor functionality and complement activity. The antibody bound NP from several CCHFV strains and exhibited robust cross-protection against the heterologous CCHFV strain Afg09-2990. Our work demonstrates that the NP is a viable target for antibody-based therapeutics, providing another direction for developing immunotherapeutics against CCHFV.
Collapse
Affiliation(s)
- Aura R Garrison
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA.
| | - Vanessa Moresco
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Curtis R Cline
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Michael D Ward
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Keersten M Ricks
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Scott P Olschner
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Lisa H Cazares
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Elif Karaaslan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Collin J Fitzpatrick
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Scott D Pegan
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
- Department of Chemistry & Life Science, United States Military Academy, West Point, NY, USA
| | - Joseph W Golden
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA.
| |
Collapse
|
3
|
Zhang P, Li M, Zhang Y, Lian C, Sun J, He Y, Hu W, Wang L, Li T, Liu S, Zhang Y. Plasma proteomic profiling reveals KRT19 could be a potential biomarker in patients with anti-MDA5+ dermatomyositis. Clin Rheumatol 2023:10.1007/s10067-023-06624-6. [PMID: 37160775 DOI: 10.1007/s10067-023-06624-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/29/2023] [Accepted: 05/03/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE To investigate the immune response-related protein profiling in plasma of patients with idiopathic inflammatory myopathies (IIMs), especially in anti-MDA5+ dermatomyositis (DM). METHODS A total of 166 IIM patients and 107 healthy controls (HCs) were enrolled in our study. Ninety-two plasma immune response-related proteins were detected by Olink proteomics in 36 IIM patients and 25 HCs. The expression of plasma KRT19 was validated in another 130 IIM patients, 82 HCs, and 55 other rheumatic diseases. RESULTS A total of 46 differentially expressed proteins were detected, including 12 upregulated proteins and 34 downregulated proteins in IIM patients compared with HCs. Pathway analysis revealed lactoferrin danger signal response pathway, TLR4 signaling and tolerance, infection, and IL-10 signaling pathway were activated. The immune response-related protein profiling significantly altered in anti-MDA5+ DM patients, with LAMP3, HSD11B1, and KRT19 significantly increased, while SH2D1A, ITGA11, TRIM21, CD28, ITGB6, and HEXIM1 tremendously decreased. In addition, KRT19 was significantly increased in IIM patients, especially in anti-MDA5+ DM patients with the diagnostic value of a significant area under the ROC curve of 0.881. CONCLUSION Immune response-related proteins are significantly altered in patients with anti-MDA5+ DM patients. KRT19 could be a potential biomarker for anti-MDA5+ DM patients. Key Points • What is already known on this topic? Anti-MDA5+ DM is a distinctive subtype of IIM. Plasma immune response-related proteins panel needs to be investigated. • What this study adds? Plasma protein profiling of immune response-related proteins significantly altered in patients with idiopathic inflammatory myopathies (IIM), especially in anti-MDA5+ DM patients. • How this study might affect research, practice, or policy? KRT19 could be a potential biomarker in patients with anti-MDA5+ dermatomyositis.
Collapse
Affiliation(s)
- Panpan Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Mengdi Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Yuqi Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Chaofeng Lian
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Jinlei Sun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Yujie He
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Wenlu Hu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Limei Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Tianfang Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Shengyun Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China.
| | - Yusheng Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China.
| |
Collapse
|
4
|
Rabdano SO, Ruzanova EA, Pletyukhina IV, Saveliev NS, Kryshen KL, Katelnikova AE, Beltyukov PP, Fakhretdinova LN, Safi AS, Rudakov GO, Arakelov SA, Andreev IV, Kofiadi IA, Khaitov MR, Valenta R, Kryuchko DS, Berzin IA, Belozerova NS, Evtushenko AE, Truhin VP, Skvortsova VI. Immunogenicity and In Vivo Protective Effects of Recombinant Nucleocapsid-Based SARS-CoV-2 Vaccine Convacell ®. Vaccines (Basel) 2023; 11:vaccines11040874. [PMID: 37112786 PMCID: PMC10141225 DOI: 10.3390/vaccines11040874] [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: 04/02/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
The vast majority of SARS-CoV-2 vaccines which are licensed or under development focus on the spike (S) protein and its receptor binding domain (RBD). However, the S protein shows considerable sequence variations among variants of concern. The aim of this study was to develop and characterize a SARS-CoV-2 vaccine targeting the highly conserved nucleocapsid (N) protein. Recombinant N protein was expressed in Escherichia coli, purified to homogeneity by chromatography and characterized by SDS-PAGE, immunoblotting, mass spectrometry, dynamic light scattering and differential scanning calorimetry. The vaccine, formulated as a squalane-based emulsion, was used to immunize Balb/c mice and NOD SCID gamma (NSG) mice engrafted with human PBMCs, rabbits and marmoset monkeys. Safety and immunogenicity of the vaccine was assessed via ELISA, cytokine titer assays and CFSE dilution assays. The protective effect of the vaccine was studied in SARS-CoV-2-infected Syrian hamsters. Immunization induced sustainable N-specific IgG responses and an N-specific mixed Th1/Th2 cytokine response. In marmoset monkeys, an N-specific CD4+/CD8+ T cell response was observed. Vaccinated Syrian hamsters showed reduced lung histopathology, lower virus proliferation, lower lung weight relative to the body, and faster body weight recovery. Convacell® thus is shown to be effective and may augment the existing armamentarium of vaccines against COVID-19.
Collapse
Affiliation(s)
- Sevastyan O Rabdano
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Ellina A Ruzanova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Iuliia V Pletyukhina
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Nikita S Saveliev
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | | | | | - Petr P Beltyukov
- Scientific Research Institute of Hygiene, Occupational Pathology and Human Ecology of the Federal Medical-Biological Agency of Russia (SRIHOPHE), Kuzmolovsky 188663, Russia
| | - Liliya N Fakhretdinova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Ariana S Safi
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - German O Rudakov
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Sergei A Arakelov
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Igor V Andreev
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
| | - Ilya A Kofiadi
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
- Department of Immunology, N.I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow 117997, Russia
| | - Musa R Khaitov
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
- Department of Immunology, N.I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow 117997, Russia
| | - Rudolf Valenta
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, Moscow 119435, Russia
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Daria S Kryuchko
- Federal Medical-Biological Agency of Russia, Moscow 125310, Russia
| | - Igor A Berzin
- Federal Medical-Biological Agency of Russia, Moscow 125310, Russia
| | - Natalia S Belozerova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Anatoly E Evtushenko
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Viktor P Truhin
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | | |
Collapse
|
5
|
Li M, Huang J, Zhu Y, Huang Z, Zhang G, Huang J. Anti-L1 antibody-bound HPV16 pseudovirus is degraded intracellularly via TRIM21/proteasomal pathway. Virol J 2022; 19:90. [PMID: 35619167 PMCID: PMC9137102 DOI: 10.1186/s12985-022-01826-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background Persistent HPV16 infection is the leading risk factor for developing cervical cancer. Anti-L1 antibodies against HPV16 produced in HPV16 infections play diverse roles in the clearance of virus infection and prevention of persistence. It has been implicated that the cervicovaginal squamous epithelial cells actually express TRIM21 and that some HPV16 particles could escape leaky endosomal compartment into the cytosol and that Fc receptor TRIM21 directly neutralize infection by targeting antibody-opsonized viruses for proteasomal degradation. We explored whether anti-L1 antibody opsonized HPV16 pseudovirus (PsV) entered into the cytosol could be neutralized by TRIM21-mediated activation of a proteasomal pathway to reduce the chance of persistent HPV16 infection. Methods HPV16 PsV were generated and extracted in HEK 293FT cells co-transfected with pcDNA3.1-eGFP and p16sheLL plasmids according to the standard protocol. The HPV16 PsV with capsid protein L1 was characterized by fluorescence microscopy and western blot, and the HPV16 PsV titer and anti-L1-bound PsV entry efficiency were detected by flow cytometry. The expressions of transcription factors (TF) and cytokines elicited by the TRIM21-activated proteasomal pathway were confirmed by dual-luciferase reporter assay and RT-qPCR. The changes in HPV16 PsV load with or without inhibitors in the infected HEK 293FT cells were determinated by qPCR. Results Simultaneous transfection with pcDNA3.1-eGFP and p16sheLL plasmids into the HEK 293FT cells resulted in the self-assembly of HPV16 PsV with capsid protein L1. Both HPV16 PsV and anti-L1-bound HPV16 PsV could infect HEK 293FT cells. Anti-L1-bound PsV up-regulated TRIM21 mediated-activation of proteasome and increased expressions of TF and cytokines in the infected cells where HPV16 PsV load reduced by ~ 1000-fold in the presence of anti-L1 antibody, but inhibition of proteasomal activity increased HPV16 PsV load. Conclusion Our preliminary results indicate that anti-L1 antibody entered with HPV16 PsV into the cells could mediate degradation of HPV16 PsV by TRIM21-activated proteasomal pathway intracellularly, giving anti-capsid protein L1 antibody a role in host defense of persistent HPV16 infection.
Collapse
Affiliation(s)
- Meiying Li
- Department of Biochemistry and Molecular Biology, Sichuan Cancer Institute, Chengdu, 610041, People's Republic of China
| | - Jianmei Huang
- Department of Gynecologic Oncology, Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| | - Yi Zhu
- Department of Gynecologic Oncology, Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China.,Department of Ultrasound, Sichuan Cancer Hospital, Chengdu, 610041, People's Republic of China
| | - Ziyi Huang
- Department of Bioinformatics, Basic Medical College of Chongqing Medical University, Chongqing, People's Republic of China
| | - Guonan Zhang
- Department of Gynecologic Oncology, Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, People's Republic of China
| | - Jianming Huang
- Department of Biochemistry and Molecular Biology, Sichuan Cancer Institute, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
6
|
Cai C, Tang YD, Zheng C. When RING Finger Family Proteins meet SARS-CoV-2. J Med Virol 2022; 94:2977-2985. [PMID: 35257387 DOI: 10.1002/jmv.27701] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
The pandemic coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is currently the most formidable challenge to humans. Understanding the complicated virus-host interplay is crucial for fighting against viral infection. A growing number of studies point to the critical roles of RING (really interesting new gene) finger (RNF) proteins during SARS-CoV-2 infection. RNF proteins exert direct antiviral activity by targeting genome and envelope glycoproteins of SARS-CoV-2. Additionally, some RNF members serve as potent regulators for antiviral innate immunity and antibody-dependent neutralization of SARS-CoV-2. Notably, SARS-CoV-2 also hijacks the RNF proteins-mediated ubiquitination process to evade host antiviral innate immunity and enhance viral replication. In this mini-review, we discuss the diverse antiviral mechanisms of RNF proteins and viral immune evasion in an RNF proteins-dependent manner. Understanding the crosstalk between RNF proteins and SARS-CoV-2 infection would help design potential novel targets for COVID-19 treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Chunmei Cai
- Research Center for High Altitude Medicine, School of Medical, Qinghai University, Xining, China.,Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Qinghai University, Xining, China
| | - Yan-Dong Tang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
7
|
Zeng J, Santos AF, Mukadam AS, Osswald M, Jacques DA, Dickson CF, McLaughlin SH, Johnson CM, Kiss L, Luptak J, Renner N, Vaysburd M, McEwan WA, Morais-de-Sá E, Clift D, James LC. Target-induced clustering activates Trim-Away of pathogens and proteins. Nat Struct Mol Biol 2021; 28:278-289. [PMID: 33633400 PMCID: PMC7611929 DOI: 10.1038/s41594-021-00560-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/13/2021] [Indexed: 01/31/2023]
Abstract
Trim-Away is a recently developed technology that exploits off-the-shelf antibodies and the RING E3 ligase and cytosolic antibody receptor TRIM21 to carry out rapid protein depletion. How TRIM21 is catalytically activated upon target engagement, either during its normal immune function or when repurposed for targeted protein degradation, is unknown. Here we show that a mechanism of target-induced clustering triggers intermolecular dimerization of the RING domain to switch on the ubiquitination activity of TRIM21 and induce virus neutralization or drive Trim-Away. We harness this mechanism for selective degradation of disease-causing huntingtin protein containing long polyglutamine tracts and expand the Trim-Away toolbox with highly active TRIM21-nanobody chimeras that can also be controlled optogenetically. This work provides a mechanism for cellular activation of TRIM RING ligases and has implications for targeted protein degradation technologies.
Collapse
Affiliation(s)
- Jingwei Zeng
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Ana Filipa Santos
- i3S - Instituto de Investigação e Inovação em Saúde and IBMC Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Aamir S. Mukadam
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mariana Osswald
- i3S - Instituto de Investigação e Inovação em Saúde and IBMC Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - David A. Jacques
- EMBL Australia Node, Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Claire F. Dickson
- EMBL Australia Node, Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | | | | - Leo Kiss
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Jakub Luptak
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Nadine Renner
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Marina Vaysburd
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - William A. McEwan
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK,Correspondence: William McEwan (); Eurico Morais-de-Sá (); Dean Clift (); Leo C. James ()
| | - Eurico Morais-de-Sá
- i3S - Instituto de Investigação e Inovação em Saúde and IBMC Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal,Correspondence: William McEwan (); Eurico Morais-de-Sá (); Dean Clift (); Leo C. James ()
| | - Dean Clift
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK,Correspondence: William McEwan (); Eurico Morais-de-Sá (); Dean Clift (); Leo C. James ()
| | - Leo C. James
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK,Correspondence: William McEwan (); Eurico Morais-de-Sá (); Dean Clift (); Leo C. James ()
| |
Collapse
|
8
|
Pozzi S, Codron P, Soucy G, Renaud L, Cordeau PJ, Dutta K, Bareil C, Julien JP. Monoclonal full-length antibody against TAR DNA binding protein 43 reduces related proteinopathy in neurons. JCI Insight 2020; 5:140420. [PMID: 33021970 PMCID: PMC7710295 DOI: 10.1172/jci.insight.140420] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), 2 incurable neurodegenerative disorders, share the same pathological hallmark named TDP43 (TAR DNA binding protein 43) proteinopathy. This event is characterized by a consistent cytoplasmic mislocalization and aggregation of the protein TDP43, which loses its physiological properties, leading neurons to death. Antibody-based approaches are now emerging interventions in the field of neurodegenerative disorders. Here, we tested the target specificity, in vivo distribution, and therapeutic efficacy of a monoclonal full-length antibody, named E6, in TDP43-related conditions. We observed that the antibody recognizes specifically the cytoplasmic fraction of TDP43. We demonstrated its ability in targeting large neurons in the spinal cord of mice and in reducing TDP43 mislocalization and NF-κB activation. We also recognized the proteasome as well as the lysosome machineries as possible mechanisms used by the antibody to reduce TDP43 proteinopathy. To our knowledge, this is the first report showing the therapeutic efficacy and feasibility of a full-length antibody against TDP43 in reducing TDP43 proteinopathy in spinal neurons of an ALS/FTLD mouse model. A full-length antibody against TDP43 reduces TDP43 proteinopathy in spinal neurons of an amyotrophic lateral sclerosis/Frontotemporal lobar degeneration mouse model.
Collapse
Affiliation(s)
- Silvia Pozzi
- CERVO Brain Research Centre, Québec, Québec, Canada
| | - Philippe Codron
- UMR CNRS 6015, INSERM U1083, University of Angers, Angers, France
| | | | | | | | - Kallol Dutta
- CERVO Brain Research Centre, Québec, Québec, Canada
| | | | - Jean-Pierre Julien
- CERVO Brain Research Centre, Québec, Québec, Canada.,Department of Psychiatry and Neuroscience, University of Laval, Québec City, Canada
| |
Collapse
|
9
|
Williams FP, Haubrich K, Perez-Borrajero C, Hennig J. Emerging RNA-binding roles in the TRIM family of ubiquitin ligases. Biol Chem 2020; 400:1443-1464. [PMID: 31120853 DOI: 10.1515/hsz-2019-0158] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022]
Abstract
TRIM proteins constitute a large, diverse and ancient protein family which play a key role in processes including cellular differentiation, autophagy, apoptosis, DNA repair, and tumour suppression. Mostly known and studied through the lens of their ubiquitination activity as E3 ligases, it has recently emerged that many of these proteins are involved in direct RNA binding through their NHL or PRY/SPRY domains. We summarise the current knowledge concerning the mechanism of RNA binding by TRIM proteins and its biological role. We discuss how RNA-binding relates to their previously described functions such as E3 ubiquitin ligase activity, and we will consider the potential role of enrichment in membrane-less organelles.
Collapse
Affiliation(s)
- Felix Preston Williams
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Kevin Haubrich
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Cecilia Perez-Borrajero
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Janosch Hennig
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany, e-mail:
| |
Collapse
|
10
|
Li Z, Huan C, Wang H, Liu Y, Liu X, Su X, Yu J, Zhao Z, Yu XF, Zheng B, Zhang W. TRIM21-mediated proteasomal degradation of SAMHD1 regulates its antiviral activity. EMBO Rep 2020; 21:e47528. [PMID: 31797533 PMCID: PMC6944907 DOI: 10.15252/embr.201847528] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 10/09/2019] [Accepted: 11/13/2019] [Indexed: 01/01/2023] Open
Abstract
SAMHD1 possesses multiple functions, but whether cellular factors regulate SAMHD1 expression or its function remains not well characterized. Here, by investigating why cultured RD and HEK293T cells show different sensitivity to enterovirus 71 (EV71) infection, we demonstrate that SAMHD1 is a restriction factor for EV71. Importantly, we identify TRIM21, an E3 ubiquitin ligase, as a key regulator of SAMHD1, which specifically interacts and degrades SAMHD1 through the proteasomal pathway. However, TRIM21 has no effect on EV71 replication itself. Moreover, we prove that interferon production stimulated by EV71 infection induces increased TRIM21 and SAMHD1 expression, whereas increasing TRIM21 overrides SAMHD1 inhibition of EV71 in cells and in a neonatal mouse model. TRIM21-mediated degradation of SAMHD1 also affects SAMHD1-dependent restriction of HIV-1 and the regulation of interferon production. We further identify the functional domains in TRIM21 required for SAMHD1 binding and the ubiquitination site K622 in SAMHD1 and show that phosphorylation of SAMHD1 at T592 also blocks EV71 restriction. Our findings illuminate how EV71 overcomes SAMHD1 inhibition via the upregulation of TRIM21.
Collapse
Affiliation(s)
- Zhaolong Li
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| | - Chen Huan
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| | - Hong Wang
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| | - Yue Liu
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| | - Xin Liu
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| | - Xing Su
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| | - Jinghua Yu
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| | - Zhilei Zhao
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| | - Xiao-Fang Yu
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Baisong Zheng
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| | - Wenyan Zhang
- The First Hospital of Jilin University, Institute of Virology and AIDS Research, Changchun, China
| |
Collapse
|
11
|
Foss S, Bottermann M, Jonsson A, Sandlie I, James LC, Andersen JT. TRIM21-From Intracellular Immunity to Therapy. Front Immunol 2019; 10:2049. [PMID: 31555278 PMCID: PMC6722209 DOI: 10.3389/fimmu.2019.02049] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022] Open
Abstract
Tripartite motif containing-21 (TRIM21) is a cytosolic ubiquitin ligase and antibody receptor that provides a last line of defense against invading viruses. It does so by acting as a sensor that intercepts antibody-coated viruses that have evaded extracellular neutralization and breached the cell membrane. Upon engagement of the Fc of antibodies bound to viruses, TRIM21 triggers a coordinated effector and signaling response that prevents viral replication while at the same time inducing an anti-viral cellular state. This dual effector function is tightly regulated by auto-ubiquitination and phosphorylation. Therapeutically, TRIM21 has been shown to be detrimental in adenovirus based gene therapy, while it may be favorably utilized to prevent tau aggregation in neurodegenerative disorders. In addition, TRIM21 may synergize with the complement system to block viral replication as well as transgene expression. TRIM21 can also be utilized as a research tool to deplete specific proteins in cells and zebrafish embryos. Here, we review our current biological understanding of TRIM21 in light of its versatile functions.
Collapse
Affiliation(s)
- Stian Foss
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway.,Department of Immunology, Centre for Immune Regulation, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Maria Bottermann
- Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Medical Research Council, Cambridge, United Kingdom
| | - Alexandra Jonsson
- Department of Immunology, Centre for Immune Regulation, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway.,Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Medical Research Council, Cambridge, United Kingdom
| | - Inger Sandlie
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway.,Department of Immunology, Centre for Immune Regulation, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Leo C James
- Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Medical Research Council, Cambridge, United Kingdom
| | - Jan Terje Andersen
- Department of Immunology, Centre for Immune Regulation, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
Xu M, Jiang Y, Wang J, Liu J, Liu C, Liu D, Yang S. Distinct variations of antibody secreting cells and memory B cells during the course of Kawasaki disease. BMC Immunol 2019; 20:16. [PMID: 31159728 PMCID: PMC6547606 DOI: 10.1186/s12865-019-0299-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/23/2019] [Indexed: 01/02/2023] Open
Abstract
Background Both antibody secreting cells (ASCs) and memory B cells are essential for the maintenance of humoral immunity. To date, limit studies have focused on the two populations in Kawasaki disease (KD). To address the status of humoral immunity during KD, our current concentration is on the variations of ASCs and memory B cells, as well as their subsets in both acute and remission stages of KD. Methods ASCs were defined as the population with high expressions of CD27 and CD38 among CD3-CD20- lymphocytes. Based on the expression of surface marker CD138 and intracellular marker IgG, ASCs were further divided into two subsets. Memory B cells were characterized by the expressions of IgD, CD27 and IgM, upon which memory B cells were further categorized into CD27 + IgD- (switched memory, Sm), CD27-IgD- (Double negative, DN) and CD27 + IgD + IgM+ (marginal zone, MZ) B cells. Collectively, six populations were analyzed using flow cytometry. The blood samples were collected from KD patients in different stages and healthy controls. Results In the acute stage, the percentages of ASCs, CD138+ ASCs, and IgG+ ASCs were significantly increased. In contrast, the percentages of memory B cells including Sm and MZ B cells were significantly decreased. Correlation analysis found ASCs positively correlated with the level of serum IgM, whereas MZ B cells not only positively correlated with the level of serum IgG, IgA, and IgM, but also positively correlated with the level of serum complement C3 and C4 and negatively correlated with the value of C-reactive protein (CRP). In the remission stage, the percentages of IgG+ ASCs and MZ B cells were significantly reduced, whereas other subsets presented heterogeneous variations. Conclusions Our study provided direct evidence that ASCs contributed to the pathogenesis of KD, and it was the first time to describe the variation of memory B cells in this disease. Among the subsets, only IgG+ ASCs presented a significant increase in the acute stage and decreased after IVIG administration, indicating the involvement of IgG+ ASCs in the inflammation of KD and also suggesting that IVIG played an inhibitory role in the expression of cytoplasmic IgG. Electronic supplementary material The online version of this article (10.1186/s12865-019-0299-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Meng Xu
- Department of Pediatric Rheumatology, Immunology, and Allergy, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yanfang Jiang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Jinghua Wang
- Department of Pediatric Rheumatology, Immunology, and Allergy, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jinxiang Liu
- Department of Pediatric Rheumatology, Immunology, and Allergy, The First Hospital of Jilin University, Changchun, 130021, China
| | - Congcong Liu
- Department of Pediatric Rheumatology, Immunology, and Allergy, The First Hospital of Jilin University, Changchun, 130021, China
| | - Deying Liu
- Department of Pediatric Rheumatology, Immunology, and Allergy, The First Hospital of Jilin University, Changchun, 130021, China.,Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Sirui Yang
- Department of Pediatric Rheumatology, Immunology, and Allergy, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
13
|
Ng PML, Kaliaperumal N, Lee CY, Chin WJ, Tan HC, Au VB, Goh AXH, Tan QW, Yeo DSG, Connolly JE, Wang CI. Enhancing Antigen Cross-Presentation in Human Monocyte-Derived Dendritic Cells by Recruiting the Intracellular Fc Receptor TRIM21. THE JOURNAL OF IMMUNOLOGY 2019; 202:2307-2319. [PMID: 30796180 DOI: 10.4049/jimmunol.1800462] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 01/31/2019] [Indexed: 01/24/2023]
Abstract
Suboptimal immune responses to pathogens contribute to chronic infections. One way to improve immune responses is to boost Ag presentation. In this study, we investigate the potential of the tripartite motif-containing 21 (TRIM21) pathway. TRIM21 is a ubiquitously expressed cytosolic protein that recognizes the Fc region of Abs. When Abs that are bound to pathogens enter the cell as immune complexes, binding of TRIM21 to Fc initiates downstream inflammatory signaling and targets the immune complexes for proteasomal degradation. In APCs, peptides generated by proteasomes are loaded onto MHC class I molecules to stimulate CD8 T cell responses, which are crucial for effective immunity to pathogens. We hypothesized that increasing the affinity between immune complexes and TRIM21 might markedly improve CD8 T cell responses to Ags processed by the TRIM21 pathway. Using phage display technology, we engineered the human IgG1 Fc to increase its affinity for TRIM21 by 100-fold. Adenovirus immune complexes with the engineered Fc induced greater maturation of human dendritic cells (DC) than immune complexes with unmodified Fc and stimulated increased Ag-specific CD8 T cell proliferation and IFN-γ release in cocultures of DC-PBMC. Thus, by increasing the affinity between Fc and TRIM21, Ags from immune complexes undergo enhanced cross-presentation on DC, leading to greater CD8 T cell responses. Our study reveals an approach that could potentially be used in vaccines to increase cytotoxic T cell responses against Ags that are targeted or delivered by Fc-modified Abs.
Collapse
Affiliation(s)
- Patricia M L Ng
- Singapore Immunology Network, Agency for Science, Technology and Research, S138648 Singapore
| | - Nivashini Kaliaperumal
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, S138673 Singapore
| | - Chia Yin Lee
- Singapore Immunology Network, Agency for Science, Technology and Research, S138648 Singapore
| | - Wen Jie Chin
- Singapore Immunology Network, Agency for Science, Technology and Research, S138648 Singapore.,School of Biological Science, Nanyang Technological University, S637551 Singapore
| | - Hwee Ching Tan
- Singapore Immunology Network, Agency for Science, Technology and Research, S138648 Singapore
| | - Veonice B Au
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, S138673 Singapore
| | - Angeline X-H Goh
- Singapore Immunology Network, Agency for Science, Technology and Research, S138648 Singapore
| | - Qiao Wen Tan
- Singapore Immunology Network, Agency for Science, Technology and Research, S138648 Singapore.,School of Life Sciences and Chemical Technology, Ngee Ann Polytechnic, S599489 Singapore; and
| | - Darren S G Yeo
- Singapore Immunology Network, Agency for Science, Technology and Research, S138648 Singapore.,School of Life Sciences and Chemical Technology, Ngee Ann Polytechnic, S599489 Singapore; and
| | - John E Connolly
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, S138673 Singapore; .,Institute of Biomedical Studies, Baylor University, Waco, TX 76712
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research, S138648 Singapore;
| |
Collapse
|
14
|
Park H, Kim M, Seo Y, Ham Y, Cho MY, Kwon MH. Cytosolic Internalization of Anti-DNA Antibodies by Human Monocytes Induces Production of Pro-inflammatory Cytokines Independently of the Tripartite Motif-Containing 21 (TRIM21)-Mediated Pathway. Front Immunol 2018; 9:2019. [PMID: 30233598 PMCID: PMC6131520 DOI: 10.3389/fimmu.2018.02019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 08/16/2018] [Indexed: 01/05/2023] Open
Abstract
Anti-DNA autoantibodies are a hallmark of systemic lupus erythematosus (SLE). A subset of anti-DNA IgG autoantibodies is cell-internalizable; thus they can enter living cells in the form of free IgG. However, the contribution made by the Fc region of internalized free-form IgG to the cytokine response has not been studied, despite the recent discovery of tripartite motif-containing 21 (TRIM21), a cytosolic Fc receptor involved in immune signaling. This study used an internalizable IgG anti-DNA antibody (3D8) to examine the cytokine responses of human monocytes to the Fc region of cytosolic free IgG. Internalization of 3D8 IgG and a 3D8 single-chain variable fragment-Fc (scFv-Fc) induced production of IL-8 and TNF-α via activation of NF-κB. By contrast, a 3D8 scFv (comprising variable domains alone) did not. This suggests Fc-dependent cytokine signaling. A 3D8 IgG-N434D mutant that is not recognized by TRIM21 induced greater production of cytokines than 3D8 IgG. Moreover the amounts of cytokines induced by 3D8 IgG in TRIM21-knockdown THP-1 cells were higher than those in control cells, indicating that cytokine signaling is not mediated by TRIM21. The results suggest the existence of a novel Fc-dependent signaling pathway that is activated upon internalization of IgG antibodies by human monocytes.
Collapse
Affiliation(s)
- Hyunjoon Park
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Minjae Kim
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Youngsil Seo
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Yeonkyoung Ham
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Mi-Young Cho
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| | - Myung-Hee Kwon
- Department of Biomedical Sciences, Graduate School, Ajou University, Yeongtong-gu, Suwon, South Korea.,Department of Microbiology, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
15
|
Kamiyama R, Yoshimi R, Takeno M, Iribe Y, Tsukahara T, Kishimoto D, Kunishita Y, Sugiyama Y, Tsuchida N, Nakano H, Minegishi K, Tamura M, Asami Y, Kirino Y, Ishigatsubo Y, Ozato K, Nakajima H. Dysfunction of TRIM21 in interferon signature of systemic lupus erythematosus. Mod Rheumatol 2018; 28:993-1003. [PMID: 29385873 DOI: 10.1080/14397595.2018.1436028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES TRIM21 is an E3 ubiquitin ligase for interferon regulatory factors (IRFs) that are involved in innate and acquired immunity. Here, we evaluated the role of TRIM21 in the interferon (IFN) signature of systemic lupus erythematosus (SLE). METHODS Twenty SLE patients and 24 healthy controls were enrolled in this study. We analyzed mRNA expression of TRIM21, type I IFN, and IFN-inducible genes in peripheral blood mononuclear cell (PBMC). The protein levels of IRFs were assessed by Western blotting in PBMCs cultured with or without MG-132. RESULTS The expression of TRIM21 mRNA and protein was significantly higher in SLE PBMCs as compared to healthy controls. There was a correlation between TRIM21 mRNA expression and SLE activities. In contrast to a negative correlation between mRNA expression level of TRIM21 and those of type I IFNs in healthy controls, we found a positive correlation between them in anti-TRIM21 antibody-positive SLE patients. Neither positive nor negative correlation was observed in the autoantibody-negative SLE patients. Western-blotting analysis revealed impaired ubiquitin-dependent proteasomal degradation of IRFs in SLE PBMCs. CONCLUSION Our study showed ubiquitin-dependent proteasomal degradation of IRFs was impaired in anti-TRIM21 antibody-dependent and -independent fashions, leading to amplification of IFN signature in SLE.
Collapse
Affiliation(s)
- Reikou Kamiyama
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Ryusuke Yoshimi
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Mitsuhiro Takeno
- b Department of Allergy and Rheumatology , Nippon Medical School Graduate School of Medicine , Tokyo , Japan
| | - Yasuhiro Iribe
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Toshinori Tsukahara
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan.,c Department of Pulmonology , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Daiga Kishimoto
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yosuke Kunishita
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yumiko Sugiyama
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Naomi Tsuchida
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Hiroto Nakano
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Kaoru Minegishi
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Maasa Tamura
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yukiko Asami
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yohei Kirino
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Yoshiaki Ishigatsubo
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| | - Keiko Ozato
- d Program in Genomics of Differentiation , NICHD, National Institutes of Health , Bethesda , MD , USA
| | - Hideaki Nakajima
- a Department of Stem Cell and Immune Regulation , Yokohama City University Graduate School of Medicine , Yokohama , Japan
| |
Collapse
|
16
|
Schmid M, Ernst P, Honegger A, Suomalainen M, Zimmermann M, Braun L, Stauffer S, Thom C, Dreier B, Eibauer M, Kipar A, Vogel V, Greber UF, Medalia O, Plückthun A. Adenoviral vector with shield and adapter increases tumor specificity and escapes liver and immune control. Nat Commun 2018; 9:450. [PMID: 29386504 PMCID: PMC5792622 DOI: 10.1038/s41467-017-02707-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/20/2017] [Indexed: 01/16/2023] Open
Abstract
Most systemic viral gene therapies have been limited by sequestration and degradation of virions, innate and adaptive immunity, and silencing of therapeutic genes within the target cells. Here we engineer a high-affinity protein coat, shielding the most commonly used vector in clinical gene therapy, human adenovirus type 5. Using electron microscopy and crystallography we demonstrate a massive coverage of the virion surface through the hexon-shielding scFv fragment, trimerized to exploit the hexon symmetry and gain avidity. The shield reduces virion clearance in the liver. When the shielded particles are equipped with adaptor proteins, the virions deliver their payload genes into human cancer cells expressing HER2 or EGFR. The combination of shield and adapter also increases viral gene delivery to xenografted tumors in vivo, reduces liver off-targeting and immune neutralization. Our study highlights the power of protein engineering for viral vectors overcoming the challenges of local and systemic viral gene therapies. Viral gene therapy can be limited by the efficacy of virion sequestration, immune responses and the silencing of genetic payloads. Here the authors engineer an advenovirus protein coat which shields the virion from the immune system while targeting cancer cells.
Collapse
Affiliation(s)
- Markus Schmid
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Patrick Ernst
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Annemarie Honegger
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Maarit Suomalainen
- Department of Molecular Life Science, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Martina Zimmermann
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Lukas Braun
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, 8093, Zurich, Switzerland
| | - Sarah Stauffer
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Cristian Thom
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Birgit Dreier
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Matthias Eibauer
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Anja Kipar
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 268, 8057, Zurich, Switzerland
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, 8093, Zurich, Switzerland
| | - Urs F Greber
- Department of Molecular Life Science, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland.,Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, 84105, Israel
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstr, 190, 8057, Zurich, Switzerland.
| |
Collapse
|
17
|
Rhodes DA, Isenberg DA. TRIM21 and the Function of Antibodies inside Cells. Trends Immunol 2017; 38:916-926. [PMID: 28807517 DOI: 10.1016/j.it.2017.07.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/28/2017] [Accepted: 07/18/2017] [Indexed: 11/26/2022]
Abstract
Therapeutic antibodies targeting disease-associated antigens are key tools in the treatment of cancer and autoimmunity. So far, therapeutic antibodies have targeted antigens that are, or are presumed to be, extracellular. A largely overlooked property of antibodies is their functional activity inside cells. The diverse literature dealing with intracellular antibodies emerged historically from studies of the properties of some autoantibodies. The identification of tripartite motif (TRIM) 21 as an intracellular Fc receptor linking cytosolic antibody recognition to the ubiquitin proteasome system brings this research into sharper focus. We review critically the research related to intracellular antibodies, link this to the TRIM21 effector mechanism, and highlight how this work is exposing the previously restricted intracellular space to the potential of therapeutic antibodies.
Collapse
Affiliation(s)
- David A Rhodes
- Department of Pathology, Immunology Division, University of Cambridge, Cambridge, UK.
| | - David A Isenberg
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| |
Collapse
|
18
|
Kennedy PJ, Oliveira C, Granja PL, Sarmento B. Monoclonal antibodies: technologies for early discovery and engineering. Crit Rev Biotechnol 2017; 38:394-408. [PMID: 28789584 DOI: 10.1080/07388551.2017.1357002] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Antibodies are essential in modern life sciences biotechnology. Their architecture and diversity allow for high specificity and affinity to a wide array of biochemicals. Combining monoclonal antibody (mAb) technology with recombinant DNA and protein expression links antibody genotype with phenotype. Yet, the ability to select and screen for high affinity binders from recombinantly-displayed, combinatorial libraries unleashes the true power of mAbs and a flood of clinical applications. The identification of novel antibodies can be accomplished by a myriad of in vitro display technologies from the proven (e.g. phage) to the emerging (e.g. mammalian cell and cell-free) based on affinity binding as well as function. Lead candidates can be further engineered for increased affinity and half-life, reduced immunogenicity and/or enhanced manufacturing, and storage capabilities. This review begins with antibody biology and how the structure and genetic machinery relate to function, diversity, and in vivo affinity maturation and follows with the general requirements of (therapeutic) antibody discovery and engineering with an emphasis on in vitro display technologies. Throughout, we highlight where antibody biology inspires technology development and where high-throughput, "big data" and in silico strategies are playing an increasing role. Antibodies dominate the growing class of targeted therapeutics, alone or as bioconjugates. However, their versatility extends to research, diagnostics, and beyond.
Collapse
Affiliation(s)
- Patrick J Kennedy
- a i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,b INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,c IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal
| | - Carla Oliveira
- a i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,c IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto , Porto , Portugal
| | - Pedro L Granja
- a i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,b INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,d ICBAS - Instituto de Ciências Biomédicas Abel Salazar , Universidade do Porto , Porto , Portugal.,e Departmento de Engenharia Metalúrgica e de Materiais , FEUP - Faculdade de Engenharia da Universidade do Porto , Porto , Portugal
| | - Bruno Sarmento
- a i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto , Porto , Portugal.,b INEB - Instituto de Engenharia Biomédica , Universidade do Porto , Porto , Portugal.,f CESPU , Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde & Instituto Universitário de Ciências da Saúde , Gandra , Portugal
| |
Collapse
|
19
|
Lenart M, Rutkowska-Zapała M, Szatanek R, Węglarczyk K, Stec M, Bukowska-Strakova K, Gruca A, Czyż J, Siedlar M. Reprint of: Alterations of TRIM21-mRNA expression during monocyte maturation. Immunobiology 2017; 222:841-845. [PMID: 28576352 DOI: 10.1016/j.imbio.2017.05.005] [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: 07/28/2016] [Revised: 10/14/2016] [Accepted: 10/17/2016] [Indexed: 10/19/2022]
Abstract
Tripartite motif-containing protein 21 (TRIM21) play a dual role in the cytoplasm of the cells where it facilitates destruction of some antibody-coated viruses and some bacteria, and initiates synthesis of proinflammatory cytokines. Macrophages and CD16+ monocyte subset can particularly participate in a proinflammatory response caused by viral infection, however, the molecular mechanisms underlying these processes are not fully understood. The aim of this study was to determine the level of TRIM21-mRNA expression in monocyte subsets including: classical (CD14++CD16-), intermediate (CD14++CD16+) and non-classical (CD14+CD16++) monocytes, as well as during in vitro differentiation of the isolated monocytes towards dendritic cells or macrophages. Our results revealed that the level of TRIM21 mRNA expression was significantly lower in CD16- monocytes, when compared to CD16+ cells and the whole monocyte population, yet no significant differences were observed when CD16+ population was divided into intermediate and non-classical subsets. More pronounced differences were observed in the case of monocyte-derived macrophages (MDM) and dendritic cells (DCs). TRIM21-mRNA expression level was app. 6-fold higher in DCs, and app. 16-fold higher in MDM (p<0,01), when compared to freshly isolated monocytes. Our results may suggest the new mechanism of increased proinflammatory cytokine production by CD16+ (intermediate and non-classical) monocytes and macrophages, at least in patients with acute or chronic infections, caused by enveloped viruses. We suggest that TRIM21 may be one of the factors associated with the "switching on" the proinflammatory programme in CD16+ monocytes or monocyte-derived macrophages.
Collapse
Affiliation(s)
- Marzena Lenart
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Rutkowska-Zapała
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Rafał Szatanek
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Małgorzata Stec
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Karolina Bukowska-Strakova
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Gruca
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Jarosław Czyż
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Paediatrics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
20
|
Alterations of TRIM21-mRNA expression during monocyte maturation. Immunobiology 2016; 222:494-498. [PMID: 27773663 DOI: 10.1016/j.imbio.2016.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/14/2016] [Accepted: 10/17/2016] [Indexed: 11/21/2022]
Abstract
Tripartite motif-containing protein 21 (TRIM21) play a dual role in the cytoplasm of the cells where it facilitates destruction of some antibody-coated viruses and some bacteria, and initiates synthesis of proinflammatory cytokines. Macrophages and CD16+ monocyte subset can particularly participate in a proinflammatory response caused by viral infection, however, the molecular mechanisms underlying these processes are not fully understood. The aim of this study was to determine the level of TRIM21-mRNA expression in monocyte subsets including: classical (CD14++CD16-), intermediate (CD14++CD16+) and non-classical (CD14+CD16++) monocytes, as well as during in vitro differentiation of the isolated monocytes towards dendritic cells or macrophages. Our results revealed that the level of TRIM21 mRNA expression was significantly lower in CD16- monocytes, when compared to CD16+ cells and the whole monocyte population, yet no significant differences were observed when CD16+ population was divided into intermediate and non-classical subsets. More pronounced differences were observed in the case of monocyte-derived macrophages (MDM) and dendritic cells (DCs). TRIM21-mRNA expression level was app. 6-fold higher in DCs, and app. 16-fold higher in MDM (p<0,01), when compared to freshly isolated monocytes. Our results may suggest the new mechanism of increased proinflammatory cytokine production by CD16+ (intermediate and non-classical) monocytes and macrophages, at least in patients with acute or chronic infections, caused by enveloped viruses. We suggest that TRIM21 may be one of the factors associated with the "switching on" the proinflammatory programme in CD16+ monocytes or monocyte-derived macrophages.
Collapse
|
21
|
Abstract
Mouse and human FcRs have been a major focus of attention not only of the scientific community, through the cloning and characterization of novel receptors, and of the medical community, through the identification of polymorphisms and linkage to disease but also of the pharmaceutical community, through the identification of FcRs as targets for therapy or engineering of Fc domains for the generation of enhanced therapeutic antibodies. The availability of knockout mouse lines for every single mouse FcR, of multiple or cell-specific--'à la carte'--FcR knockouts and the increasing generation of hFcR transgenics enable powerful in vivo approaches for the study of mouse and human FcR biology. This review will present the landscape of the current FcR family, their effector functions and the in vivo models at hand to study them. These in vivo models were recently instrumental in re-defining the properties and effector functions of FcRs that had been overlooked or discarded from previous analyses. A particular focus will be made on the (mis)concepts on the role of high-affinity IgG receptors in vivo and on results from antibody engineering to enhance or abrogate antibody effector functions mediated by FcRs.
Collapse
Affiliation(s)
- Pierre Bruhns
- Unité des Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur, Paris, France.,INSERM, U760, Paris, France
| | - Friederike Jönsson
- Unité des Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur, Paris, France.,INSERM, U760, Paris, France
| |
Collapse
|
22
|
Abstract
IgG4, the least represented human IgG subclass in serum, is an intriguing antibody with unique biological properties, such as the ability to undergo Fab-arm exchange and limit immune complex formation. The lack of effector functions, such as antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity, is desirable for therapeutic purposes. IgG4 plays a protective role in allergy by acting as a blocking antibody, and inhibiting mast cell degranulation, but a deleterious role in malignant melanoma, by impeding IgG1-mediated anti-tumor immunity. These findings highlight the importance of understanding the interaction between IgG4 and Fcγ receptors. Despite a wealth of structural information for the IgG1 subclass, including complexes with Fcγ receptors, and structures for intact antibodies, high-resolution crystal structures were not reported for IgG4-Fc until recently. Here, we highlight some of the biological properties of human IgG4, and review the recent crystal structures of IgG4-Fc. We discuss the unexpected conformations adopted by functionally important Cγ2 domain loops, and speculate about potential implications for the interaction between IgG4 and FcγRs.
Collapse
Affiliation(s)
- Anna M Davies
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.,Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Brian J Sutton
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.,Medical Research Council & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| |
Collapse
|
23
|
Foss S, Watkinson R, Sandlie I, James LC, Andersen JT. TRIM21: a cytosolic Fc receptor with broad antibody isotype specificity. Immunol Rev 2016; 268:328-39. [PMID: 26497531 PMCID: PMC4670481 DOI: 10.1111/imr.12363] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Antibodies are key molecules in the fight against infections. Although previously thought to mediate protection solely in the extracellular environment, recent research has revealed that antibody-mediated protection extends to the cytosolic compartment of cells. This postentry viral defense mechanism requires binding of the antibody to a cytosolic Fc receptor named tripartite motif containing 21 (TRIM21). In contrast to other Fc receptors, TRIM21 shows remarkably broad isotype specificity as it does not only bind IgG but also IgM and IgA. When viral pathogens coated with these antibody isotypes enter the cytosol, TRIM21 is rapidly recruited and efficient neutralization occurs before the virus has had the time to replicate. In addition, inflammatory signaling is induced. As such, TRIM21 acts as a cytosolic sensor that engages antibodies that have failed to protect against infection in the extracellular environment. Here, we summarize our current understanding of how TRIM21 orchestrates humoral immunity in the cytosolic environment.
Collapse
Affiliation(s)
- Stian Foss
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, Oslo, Norway.,CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
| | - Ruth Watkinson
- Protein and Nucleic Acid Chemistry Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Inger Sandlie
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, Oslo, Norway.,CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
| | - Leo C James
- Protein and Nucleic Acid Chemistry Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Jan Terje Andersen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
Affiliation(s)
- David A. Rhodes
- Department of Pathology, Immunology Division, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom; ,
| | - Walter Reith
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, CH-1211 Geneva 4, Switzerland;
| | - John Trowsdale
- Department of Pathology, Immunology Division, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom; ,
| |
Collapse
|
25
|
Fan W, Zhang D, Qian P, Qian S, Wu M, Chen H, Li X. Swine TRIM21 restricts FMDV infection via an intracellular neutralization mechanism. Antiviral Res 2016; 127:32-40. [PMID: 26777733 DOI: 10.1016/j.antiviral.2016.01.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 01/06/2016] [Accepted: 01/13/2016] [Indexed: 12/24/2022]
Abstract
The tripartite motif protein 21 (TRIM21) is a ubiquitously expressed E3 ubiquitin ligase and an intracellular antibody receptor. TRIM21 mediates antibody-dependent intracellular neutralization (ADIN) in cytosol and provides an intracellular immune response to protect host defense against pathogen infection. In this study, swine TRIM21 (sTRIM21) was cloned and its role in ADIN was investigated. The expression of sTRIM21 is induced by type I interferon in PK-15 cells. sTRIM21 restricts FMDV infection in the presence of FMDV specific antibodies. Furthermore, sTRIM21 interacts with Fc fragment of swine immunoglobulin G (sFc) fused VP1 of FMDV and thereby causing its degradation. Both the RING and SPRY domains are essential for sTRIM21 to degrade sFc-fused VP1. These results suggest that the intracellular neutralization features of FMDV contribute to the antiviral activity of sTRIM21. sTRIM21 provide another intracellular mechanism to inhibit FMDV infection in infected cells.
Collapse
Affiliation(s)
- Wenchun Fan
- State Key Laboratory of Agriculture Microbiology, Huazhong Agricultural University, Wuhan 430070, PR China; Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Dong Zhang
- State Key Laboratory of Agriculture Microbiology, Huazhong Agricultural University, Wuhan 430070, PR China; Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ping Qian
- State Key Laboratory of Agriculture Microbiology, Huazhong Agricultural University, Wuhan 430070, PR China; Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Suhong Qian
- State Key Laboratory of Agriculture Microbiology, Huazhong Agricultural University, Wuhan 430070, PR China; Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Mengge Wu
- State Key Laboratory of Agriculture Microbiology, Huazhong Agricultural University, Wuhan 430070, PR China; Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Huanchun Chen
- State Key Laboratory of Agriculture Microbiology, Huazhong Agricultural University, Wuhan 430070, PR China; Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xiangmin Li
- State Key Laboratory of Agriculture Microbiology, Huazhong Agricultural University, Wuhan 430070, PR China; Division of Animal Infectious Diseases, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China.
| |
Collapse
|
26
|
Pleass RJ, Moore SC, Stevenson L, Hviid L. Immunoglobulin M: Restrainer of Inflammation and Mediator of Immune Evasion by Plasmodium falciparum Malaria. Trends Parasitol 2015; 32:108-119. [PMID: 26597020 DOI: 10.1016/j.pt.2015.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/16/2015] [Accepted: 09/23/2015] [Indexed: 02/06/2023]
Abstract
Immunoglobulin M (IgM) is an ancient antibody class that is found in all vertebrates, with the exception of coelacanths, and is indispensable in both innate and adaptive immunity. The equally ancient human malaria parasite, Plasmodium falciparum, formed an intimate relationship with IgM with which it co-evolved. In this article, we discuss the association between IgM and human malaria parasites, building on several recent publications that implicate IgM as a crucial molecule that determines both host and parasite survival. Consequently, a better understanding of this association may lead to the development of improved intervention strategies.
Collapse
Affiliation(s)
- Richard J Pleass
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| | - Shona C Moore
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK; Warwick Systems Biology Centre, Senate House, University of Warwick, Coventry, CV4 7AL, UK
| | - Liz Stevenson
- Centre for Medical Parasitology, Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Lars Hviid
- Centre for Medical Parasitology, Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark.
| |
Collapse
|
27
|
Chan KR, Ong EZ, Mok DZL, Ooi EE. Fc receptors and their influence on efficacy of therapeutic antibodies for treatment of viral diseases. Expert Rev Anti Infect Ther 2015; 13:1351-60. [PMID: 26466016 PMCID: PMC4673539 DOI: 10.1586/14787210.2015.1079127] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The lack of vaccines against several important viral diseases necessitates the development of therapeutics to save lives and control epidemics. In recent years, therapeutic antibodies have received considerable attention due to their good safety profiles and clinical success when used against viruses such as respiratory syncytial virus, Ebola virus and Hendra virus. The binding affinity of these antibodies can directly impact their therapeutic efficacy. However, we and others have also demonstrated that the subtype of Fc-gamma receptors (FcγRs) engaged influences the stoichiometric requirement for virus neutralization. Hence, the development of therapeutic antibodies against infectious diseases should consider the FcγRs engaged and Fc-effector functions involved. This review highlights the current state of knowledge about FcγRs and FcγR effector functions involved in virus neutralization, with emphasis on factors that can affect FcγR engagement. A better understanding of Fc-FcγR interactions during virus neutralization will allow development of therapeutic antibodies that are efficacious and can be administered with minimal side effects.
Collapse
Affiliation(s)
- Kuan Rong Chan
- a 1 Program in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Eugenia Z Ong
- b 2 Experimental Therapeutics Centre, Agency for Science Technology and Research, 31 Biopolis Way, Singapore 138669, Singapore
| | - Darren Z L Mok
- c 3 Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Eng Eong Ooi
- a 1 Program in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| |
Collapse
|
28
|
Hanson QM, Barb AW. A perspective on the structure and receptor binding properties of immunoglobulin G Fc. Biochemistry 2015; 54:2931-42. [PMID: 25926001 DOI: 10.1021/acs.biochem.5b00299] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Recombinant antibodies spurred a revolution in medicine that saw the introduction of powerful therapeutics for treating a wide range of diseases, from cancers to autoimmune disorders and transplant rejection, with more applications looming on the horizon. Many of these therapeutic monoclonal antibodies (mAbs) are based on human immunoglobulin G1 (IgG1) or contain at least a portion of the molecule. Most mAbs require interactions with cell surface receptors for efficacy, including the Fc γ receptors. High-resolution structural models of antibodies and antibody fragments have been available for nearly 40 years; however, a thorough description of the structural features that determine the affinity with which antibodies interact with human receptors has not been published. In this review, we will cover the relevant history of IgG-related literature and how recent developments have changed our view of critical antibody-cell interactions at the atomic level with a nod to outstanding questions in the field and future prospects.
Collapse
Affiliation(s)
- Quinlin M Hanson
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, 2214 Molecular Biology Building, Ames, Iowa 50011, United States
| | - Adam W Barb
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, 2214 Molecular Biology Building, Ames, Iowa 50011, United States
| |
Collapse
|
29
|
McEwan WA, James LC. TRIM21-Dependent Intracellular Antibody Neutralization of Virus Infection. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 129:167-87. [DOI: 10.1016/bs.pmbts.2014.10.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
30
|
Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol 2014; 5:520. [PMID: 25368619 PMCID: PMC4202688 DOI: 10.3389/fimmu.2014.00520] [Citation(s) in RCA: 1691] [Impact Index Per Article: 169.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 10/06/2014] [Indexed: 12/21/2022] Open
Abstract
Of the five immunoglobulin isotypes, immunoglobulin G (IgG) is most abundant in human serum. The four subclasses, IgG1, IgG2, IgG3, and IgG4, which are highly conserved, differ in their constant region, particularly in their hinges and upper CH2 domains. These regions are involved in binding to both IgG-Fc receptors (FcγR) and C1q. As a result, the different subclasses have different effector functions, both in terms of triggering FcγR-expressing cells, resulting in phagocytosis or antibody-dependent cell-mediated cytotoxicity, and activating complement. The Fc-regions also contain a binding epitope for the neonatal Fc receptor (FcRn), responsible for the extended half-life, placental transport, and bidirectional transport of IgG to mucosal surfaces. However, FcRn is also expressed in myeloid cells, where it participates in both phagocytosis and antigen presentation together with classical FcγR and complement. How these properties, IgG-polymorphisms and post-translational modification of the antibodies in the form of glycosylation, affect IgG-function will be the focus of the current review.
Collapse
Affiliation(s)
- Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| | - Gillian Dekkers
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| |
Collapse
|
31
|
D'Cruz AA, Babon JJ, Norton RS, Nicola NA, Nicholson SE. Structure and function of the SPRY/B30.2 domain proteins involved in innate immunity. Protein Sci 2014; 22:1-10. [PMID: 23139046 DOI: 10.1002/pro.2185] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 10/16/2012] [Accepted: 10/19/2012] [Indexed: 11/12/2022]
Abstract
The SPRY domain is a protein interaction module found in 77 murine and ~100 human proteins, and is implicated in important biological pathways, including those that regulate innate and adaptive immunity. The current definition of the SPRY domain is based on a sequence repeat discovered in the splA kinase and ryanodine receptors. The greater SPRY family is divided into the B30.2 (which contains a PRY extension at the N-terminus) and "SPRY-only" sub-families. In this brief review, we examine the current structural and biochemical literature on SPRY/B30.2 domain involvement in key immune processes and highlight a PRY-like 60 amino acid region in the N-terminus of "SPRY-only" proteins. Phylogenetic, structural, and functional analyses suggest that this N-terminal region is related to the PRY region of B30.2 and should be characterized as part of an extended SPRY domain. Greater understanding of the functional importance of the N-terminal region in "SPRY only" proteins will enhance our ability to interrogate SPRY interactions with their respective binding partners.
Collapse
Affiliation(s)
- Akshay A D'Cruz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | | | | | | | | |
Collapse
|
32
|
Neutralization of Virus Infectivity by Antibodies: Old Problems in New Perspectives. ACTA ACUST UNITED AC 2014; 2014. [PMID: 27099867 DOI: 10.1155/2014/157895] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neutralizing antibodies (NAbs) can be both sufficient and necessary for protection against viral infections, although they sometimes act in concert with cellular immunity. Successful vaccines against viruses induce NAbs but vaccine candidates against some major viral pathogens, including HIV-1, have failed to induce potent and effective such responses. Theories of how antibodies neutralize virus infectivity have been formulated and experimentally tested since the 1930s; and controversies about the mechanistic and quantitative bases for neutralization have continually arisen. Soluble versions of native oligomeric viral proteins that mimic the functional targets of neutralizing antibodies now allow the measurement of the relevant affinities of NAbs. Thereby the neutralizing occupancies on virions can be estimated and related to the potency of the NAbs. Furthermore, the kinetics and stoichiometry of NAb binding can be compared with neutralizing efficacy. Recently, the fundamental discovery that the intracellular factor TRIM21 determines the degree of neutralization of adenovirus has provided new mechanistic and quantitative insights. Since TRIM21 resides in the cytoplasm, it would not affect the neutralization of enveloped viruses, but its range of activity against naked viruses will be important to uncover. These developments bring together the old problems of virus neutralization-mechanism, stoichiometry, kinetics, and efficacy-from surprising new angles.
Collapse
|
33
|
Verma N, Thaventhiran A, Gathmann B, Thaventhiran J, Grimbacher B. Therapeutic management of primary immunodeficiency in older patients. Drugs Aging 2014; 30:503-12. [PMID: 23605785 DOI: 10.1007/s40266-013-0079-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Primary immunodeficiency disease (PID) has traditionally been viewed as a group of illnesses seen in the paediatric age group. New advances in diagnosis and treatment have led to an increase in the number of elderly PID patients. However, there is lack of research evidence on which to base clinical management in this group of patients. Management decisions often have to be based therefore on extrapolations from other patient cohorts or from younger patients. Data from the European Society for Immunodeficiencies demonstrates that the vast majority of elderly patients suffer from predominantly antibody deficiency syndromes. We review the management of PID disease in the elderly, with a focus on antibody deficiency disease.
Collapse
Affiliation(s)
- Nisha Verma
- Department of Immunology and Molecular Pathology, Royal Free Hospital and University College London, London, UK
| | | | | | | | | | | |
Collapse
|
34
|
Su B, Moog C. Which Antibody Functions are Important for an HIV Vaccine? Front Immunol 2014; 5:289. [PMID: 24995008 PMCID: PMC4062070 DOI: 10.3389/fimmu.2014.00289] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/03/2014] [Indexed: 01/18/2023] Open
Abstract
HIV antibody (Ab) functions capable of preventing mucosal cell-free or cell-to-cell HIV transmission are critical for the development of effective prophylactic and therapeutic vaccines. In addition to CD4(+) T cells, other potential HIV-target cell types including antigen-presenting cells (APCs) (dendritic cells, macrophages) residing at mucosal sites are infected. Moreover, the interactions between APCs and HIV lead to HIV cell-to-cell transmission. Recently discovered broadly neutralizing antibodies (NAbs) are able to neutralize a broad spectrum of HIV strains, inhibit cell-to-cell transfer, and efficiently protect from infection in the experimentally challenged macaque model. However, the 31% protection observed in the RV144 vaccine trial in the absence of detectable NAbs in blood samples pointed to the possible role of additional Ab inhibitory functions. Increasing evidence suggests that IgG Fcγ receptor (FcγR)-mediated inhibition of Abs present at the mucosal site may play a role in protection against HIV mucosal transmission. Moreover, mucosal IgA Abs may be determinant in protection against HIV sexual transmission. Therefore, defining Ab inhibitory functions that could lead to protection is critical for further HIV vaccine design. Here, we review different inhibitory properties of HIV-specific Abs and discuss their potential role in protection against HIV sexual transmission.
Collapse
Affiliation(s)
- Bin Su
- INSERM U1109, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg , Strasbourg , France
| | - Christiane Moog
- INSERM U1109, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg , Strasbourg , France
| |
Collapse
|
35
|
Watkinson RE, McEwan WA, James LC. Intracellular Antibody Immunity. J Clin Immunol 2014; 34 Suppl 1:S30-4. [DOI: 10.1007/s10875-014-0017-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 03/19/2014] [Indexed: 11/29/2022]
|
36
|
Guilliams M, Bruhns P, Saeys Y, Hammad H, Lambrecht BN. The function of Fcγ receptors in dendritic cells and macrophages. Nat Rev Immunol 2014; 14:94-108. [PMID: 24445665 DOI: 10.1038/nri3582] [Citation(s) in RCA: 477] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs) and macrophages use various receptors to recognize foreign antigens and to receive feedback control from adaptive immune cells. Although it was long believed that all immunoglobulin Fc receptors are universally expressed by phagocytes, recent findings indicate that only monocyte-derived DCs and macrophages express high levels of activating Fc receptors for IgG (FcγRs), whereas conventional and plasmacytoid DCs express the inhibitory FcγR. In this Review, we discuss how the uptake, processing and presentation of antigens by DCs and macrophages is influenced by FcγR recognition of immunoglobulins and immune complexes in the steady state and during inflammation.
Collapse
Affiliation(s)
- Martin Guilliams
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Pierre Bruhns
- 1] Institut Pasteur, Département d'Immunologie, Laboratoire Anticorps en Thérapie et Pathologie, 75015 Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U760, 75015 Paris, France
| | - Yvan Saeys
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Hamida Hammad
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium. [3] Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| |
Collapse
|
37
|
Abstract
Immunotherapies targeting tau in mouse models of human tauopathies could have disease-modifying effects. In this issue of Neuron, Yanamandra et al. (2013) use tau antibodies, which effectively block tau seeding in culture, to attenuate tauopathy and improve cognition in mutant tau mouse models.
Collapse
Affiliation(s)
- Todd E Golde
- Departments of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, FL 32610, USA.
| | | | | |
Collapse
|
38
|
Abstract
Perhaps more definitively than any other class of novel Alzheimer’s disease (AD) therapy, pre-clinical studies in mouse models of amyloid β (Aβ) deposition have established the disease-modifying potential of anti-Aβ immunotherapy. Despite disappointing results to date from anti-Aβ immunotherapy therapeutic trials, there is continued hope that such immunotherapies, especially if used in the preclinical stages, could prove to be the first disease-modifying therapies available for AD. The general optimism that Aβ-targeting and emerging tau-targeting immunotherapies may prove to be disease modifying is tempered by many unanswered questions regarding these therapeutic approaches, including but not limited to i) lack of precise understanding of mechanisms of action, ii) the factors that regulate antibody exposure in the brain, iii) the optimal target epitope, and iv) the mechanisms underlying side effects. In this review I discuss how answering these and other questions could increase the likelihood of therapeutic success. As passive immunotherapies are also likely to be extremely expensive, I also raise questions relating to cost-benefit of biologic-based therapies for AD that could limit future impact of these therapies by limiting access due to economic constraints.
Collapse
Affiliation(s)
- Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
39
|
Leng L, Xu C, Wei C, Zhang J, Liu B, Ma J, Li N, Qin W, Zhang W, Zhang C, Xing X, Zhai L, Yang F, Li M, Jin C, Yuan Y, Xu P, Qin J, Xie H, He F, Wang J. A Proteomics Strategy for the Identification of FAT10-Modified Sites by Mass Spectrometry. J Proteome Res 2013; 13:268-76. [PMID: 23862649 DOI: 10.1021/pr400395k] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ling Leng
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Changming Xu
- Department of Automatic Control,
College of Mechatronics and Automation, National University of Defense Technology, Changsha 410073, China
| | - Chao Wei
- Institute of Basic Medical Science,
Chinese Academy of Medical Science and School of Basic Medicine, Peking Union Medical College, Beijing 10005, China
| | - Jiyang Zhang
- Department of Automatic Control,
College of Mechatronics and Automation, National University of Defense Technology, Changsha 410073, China
| | - Boya Liu
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Jie Ma
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Ning Li
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Weijie Qin
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Wanjun Zhang
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Chengpu Zhang
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Xiaohua Xing
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Linhui Zhai
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Fan Yang
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Mansheng Li
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Chaozhi Jin
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Yanzhi Yuan
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Ping Xu
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Jun Qin
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
- Baylor College of Medicine, Houston, Texas 77030, United States
| | - Hongwei Xie
- Department of Automatic Control,
College of Mechatronics and Automation, National University of Defense Technology, Changsha 410073, China
| | - Fuchu He
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| | - Jian Wang
- State Key Laboratory of Proteomics,
Beijing Proteome Research Center, National Center for Protein Sciences
Beijing, Beijing Institute of Radiation Medicine, Beijing 102206, China
- National Engineering Research Center for Protein Drugs, Beijing 102206, China
| |
Collapse
|
40
|
Intracellular antibody receptor TRIM21 prevents fatal viral infection. Proc Natl Acad Sci U S A 2013; 110:12397-401. [PMID: 23840060 DOI: 10.1073/pnas.1301918110] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Host species have evolved mechanisms that can inhibit pathogen replication even after a cell has been successfully invaded. Here we show that tripartite-motif protein 21 (TRIM21), a ubiquitously expressed E3 ubiquitin ligase that targets viruses inside the cytosol, protects mice against fatal viral infection. Upon infection with mouse adenovirus-1, naive mice lacking TRIM21 succumb to encephalomyelitis within 7 d. In contrast, wild-type mice rapidly up-regulate TRIM21 and control viremia. Trim21 heterozygous mice have a haploinsufficiency phenotype in which reduced TRIM21 expression leads to a viral load that is higher than wild types but lower than knockouts. TRIM21 is a high-affinity antibody receptor that allows antibodies to operate inside an infected cell. In passive transfer experiments at high viral dose, antisera that fully protects wild-type mice fails to protect most Trim21 knockout animals. These results demonstrate that TRIM21 provides potent antiviral protection and forms an important part of the humoral immune response.
Collapse
|
41
|
McEwan WA, Tam JCH, Watkinson RE, Bidgood SR, Mallery DL, James LC. Intracellular antibody-bound pathogens stimulate immune signaling via the Fc receptor TRIM21. Nat Immunol 2013; 14:327-36. [PMID: 23455675 PMCID: PMC3672961 DOI: 10.1038/ni.2548] [Citation(s) in RCA: 283] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 01/17/2013] [Indexed: 12/22/2022]
Abstract
Antibodies can be carried into the cell during pathogen infection where they are detected by the ubiquitously expressed cytosolic antibody receptor TRIM21. Here we show that TRIM21 recognition of intracellular antibodies activates immune signaling. TRIM21 catalyses K63-ubiquitin chain formation, stimulating transcription factor pathways NF-κB, AP-1 and IRF3, IRF5, IRF7. Activation results in proinflammatory cytokine production, modulation of natural killer (NK) stress ligands and the induction of an antiviral state. Intracellular antibody signaling is abrogated by genetic deletion of TRIM21 and is recovered by ectopic TRIM21 expression. Antibody sensing by TRIM21 can be stimulated upon infection by DNA or RNA non-enveloped viruses or intracellular bacteria. The antibody-TRIM21 detection system provides potent, comprehensive innate immune activation, independent of known pattern recognition receptors.
Collapse
Affiliation(s)
- William A McEwan
- Division of Protein and Nucleic Acid Chemistry, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| | | | | | | | | | | |
Collapse
|
42
|
Caoili SEC. Antidotes, antibody-mediated immunity and the future of pharmaceutical product development. Hum Vaccin Immunother 2013; 9:294-9. [PMID: 23291934 PMCID: PMC3859750 DOI: 10.4161/hv.22858] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
If new scientific knowledge is to be more efficiently generated and applied toward the advancement of health, human safety must be more effectively addressed in the conduct of research. Given the present difficulties of accurately predicting biological outcomes of novel interventions in vivo, the imperative of human safety suggests the development of novel pharmaceutical products in tandem with their prospective antidotes in anticipation of possible adverse events, to render the risks of initial clinical trials more acceptable from a regulatory standpoint. Antibody-mediated immunity provides a generally applicable mechanistic basis for developing antidotes to both biologicals and small-molecule drugs (such that antibodies may serve as antidotes to pharmaceutical agents as a class including other antibodies) and also for the control and prevention of both infectious and noninfectious diseases via passive or active immunization. Accordingly, the development of prophylactic or therapeutic passive-immunization strategies using antipeptide antibodies is a plausible prelude to the development of corresponding active-immunization strategies using peptide-based vaccines. In line with this scheme, global proliferation of antibody- and vaccine-production technologies, especially those that obviate dependence on the cold chain for storage and transport of finished products, could provide geographically distributed breakout capability against emerging and future health challenges.
Collapse
Affiliation(s)
- Salvador Eugenio C Caoili
- Department of Biochemistry and Molecular Biology; College of Medicine; University of the Philippines Manila; Manila, Philippines
| |
Collapse
|
43
|
AAA ATPase p97/VCP is essential for TRIM21-mediated virus neutralization. Proc Natl Acad Sci U S A 2012; 109:19733-8. [PMID: 23091005 DOI: 10.1073/pnas.1210659109] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tripartite motif-containing 21 (TRIM21) is a cytosolic IgG receptor that mediates intracellular virus neutralization by antibody. TRIM21 targets virions for destruction in the proteasome, but it is unclear how a substrate as large as a viral capsid is degraded. Here, we identify the ATPase p97/valosin-containing protein (VCP), an enzyme with segregase and unfoldase activity, as a key player in this process. Depletion or catalytic inhibition of VCP prevents capsid degradation and reduces neutralization. VCP is required concurrently with the proteasome, as addition of inhibitor after proteasomal degradation has no effect. Moreover, our results suggest that it is the challenging nature of virus as a substrate that necessitates involvement of VCP, since intracellularly expressed IgG Fc is degraded in a VCP-independent manner. These results implicate VCP as an important host factor in antiviral immunity.
Collapse
|
44
|
Arentz G, Thurgood LA, Lindop R, Chataway TK, Gordon TP. Secreted human Ro52 autoantibody proteomes express a restricted set of public clonotypes. J Autoimmun 2012; 39:466-70. [PMID: 22871259 DOI: 10.1016/j.jaut.2012.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 07/10/2012] [Accepted: 07/11/2012] [Indexed: 11/26/2022]
Abstract
Long-lived secreted autoantibody responses in systemic autoimmunity are generally regarded to be polyclonal and to express a diverse B-cell repertoire. Here, we have used a proteomic approach based on de novo sequencing to determine the clonality and V region structures of human autoantibodies directed against a prototypic systemic autoantigen, Ro52 (TRIM21). Remarkably, anti-Ro52 autoantibodies from patients with primary Sjögren's syndrome, systemic lupus erythematosus, systemic sclerosis or polymyositis were restricted to two IgG1 kappa clonotypes that migrated as a single species on isoelectric focusing; shared a common light chain paired with one of two closely-related heavy chains; and were public in unrelated patients. Targeted mass spectrometry using these uniquely mutated V region peptides as surrogates detected anti-Ro52 autoantibodies in human sera with high sensitivity and specificity compared with traditional ELISA. Mass spectrometry-based detection of specific autoantibody motifs provides a powerful new tool for analysis of humoral autoimmunity.
Collapse
Affiliation(s)
- Georgia Arentz
- Department of Immunology, Flinders Medical Centre and Flinders University, SA Pathology, Flinders Drive, Bedford Park, 5042 South Australia, Australia
| | | | | | | | | |
Collapse
|
45
|
Czajkowsky DM, Hu J, Shao Z, Pleass RJ. Fc-fusion proteins: new developments and future perspectives. EMBO Mol Med 2012; 4:1015-28. [PMID: 22837174 PMCID: PMC3491832 DOI: 10.1002/emmm.201201379] [Citation(s) in RCA: 336] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 05/29/2012] [Accepted: 06/15/2012] [Indexed: 12/25/2022] Open
Abstract
Since the first description in 1989 of CD4-Fc-fusion antagonists that inhibit human immune deficiency virus entry into T cells, Fc-fusion proteins have been intensely investigated for their effectiveness to curb a range of pathologies, with several notable recent successes coming to market. These promising outcomes have stimulated the development of novel approaches to improve their efficacy and safety, while also broadening their clinical remit to other uses such as vaccines and intravenous immunoglobulin therapy. This increased attention has also led to non-clinical applications of Fc-fusions, such as affinity reagents in microarray devices. Here we discuss recent results and more generally applicable strategies to improve Fc-fusion proteins for each application, with particular attention to the newer, less charted areas.
Collapse
Affiliation(s)
- Daniel M Czajkowsky
- Key Laboratory of Systems Biomedicine (Ministry of Education) & State Key Laboratory of Oncogenes & Related Genes, Shanghai Jiao Tong University, Shanghai, P. R. China
| | | | | | | |
Collapse
|
46
|
Autoantigen TRIM21/Ro52 as a Possible Target for Treatment of Systemic Lupus Erythematosus. Int J Rheumatol 2012; 2012:718237. [PMID: 22701487 PMCID: PMC3373075 DOI: 10.1155/2012/718237] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 04/01/2012] [Accepted: 04/02/2012] [Indexed: 11/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic, systemic, and autoimmune disease, whose etiology is still unknown. Although there has been progress in the treatment of SLE through the use of glucocorticoid and immunosuppressive drugs, these drugs have limited efficacy and pose significant risks of toxicity. Moreover, prognosis of patients with SLE has remained difficult to assess. TRIM21/Ro52/SS-A1, a 52-kDa protein, is an autoantigen recognized by antibodies in sera of patients with SLE and Sjögren's syndrome (SS), another systemic autoimmune disease, and anti-TRIM21 antibodies have been used as a diagnostic marker for decades. TRIM21 belongs to the tripartite motif-containing (TRIM) super family, which has been found to play important roles in innate and acquired immunity. Recently, TRIM21 has been shown to be involved in both physiological immune responses and pathological autoimmune processes. For example, TRIM21 ubiquitylates proteins of the interferon-regulatory factor (IRF) family and regulates type I interferon and proinflammatory cytokines. In this paper, we summarize molecular features of TRIM21 revealed so far and discuss its potential as an attractive therapeutic target for SLE.
Collapse
|
47
|
Current world literature. Curr Opin Rheumatol 2012; 24:435-40. [PMID: 22653148 DOI: 10.1097/bor.0b013e3283556515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
48
|
Abstract
Despite a central role in immunity, antibody neutralization of virus infection is poorly understood. Here we show how the neutralization and persistence of adenovirus type 5, a prevalent nonenveloped human virus, are dependent upon the intracellular antibody receptor TRIM21. Cells with insufficient amounts of TRIM21 are readily infected, even at saturating concentrations of neutralizing antibody. Conversely, high TRIM21 expression levels decrease the persistent fraction of the infecting virus and allows neutralization by as few as 1.6 antibody molecules per virus. The direct interaction between TRIM21 and neutralizing antibody is essential, as single-point mutations within the TRIM21-binding site in the Fc region of a potently neutralizing antibody impair neutralization. However, infection at high multiplicity can saturate TRIM21 and overcome neutralization. These results provide insight into the mechanism and importance of a newly discovered, effector-driven process of antibody neutralization of nonenveloped viruses.
Collapse
|