1
|
Zheng C, Shi Y, Zou Y. T cell co-stimulatory and co-inhibitory pathways in atopic dermatitis. Front Immunol 2023; 14:1081999. [PMID: 36993982 PMCID: PMC10040887 DOI: 10.3389/fimmu.2023.1081999] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) targeting the T cell inhibitory pathways has revolutionized cancer treatment. However, ICIs might induce progressive atopic dermatitis (AD) by affecting T cell reactivation. The critical role of T cells in AD pathogenesis is widely known. T cell co-signaling pathways regulate T cell activation, where co-signaling molecules are essential for determining the magnitude of the T cell response to antigens. Given the increasing use of ICIs in cancer treatment, a timely overview of the role of T cell co-signaling molecules in AD is required. In this review, we emphasize the importance of these molecules involved in AD pathogenesis. We also discuss the potential of targeting T cell co-signaling pathways to treat AD and present the unresolved issues and existing limitations. A better understanding of the T cell co-signaling pathways would aid investigation of the mechanism, prognosis evaluation, and treatment of AD.
Collapse
Affiliation(s)
- Chunjiao Zheng
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuling Shi
- Institute of Psoriasis, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yuling Shi, ; Ying Zou,
| | - Ying Zou
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yuling Shi, ; Ying Zou,
| |
Collapse
|
2
|
Redmond AK, Pettinello R, Bakke FK, Dooley H. Sharks Provide Evidence for a Highly Complex TNFSF Repertoire in the Jawed Vertebrate Ancestor. THE JOURNAL OF IMMUNOLOGY 2022; 209:1713-1723. [DOI: 10.4049/jimmunol.2200300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/19/2022] [Indexed: 01/04/2023]
Abstract
Abstract
Cytokines of the TNF superfamily (TNFSF) control many immunological processes and are implicated in the etiology of many immune disorders and diseases. Despite their obvious biological importance, the TNFSF repertoires of many species remain poorly characterized. In this study, we perform detailed bioinformatic, phylogenetic, and syntenic analyses of five cartilaginous fish genomes to identify their TNFSF repertoires. Strikingly, we find that shark genomes harbor ∼30 TNFSF genes, more than any other vertebrate examined to date and substantially more than humans. This is due to better retention of the ancestral jawed vertebrate TNFSF repertoire than any other jawed vertebrate lineage, combined with lineage-specific gene family expansions. All human TNFSFs appear in shark genomes, except for lymphotoxin-α (LTA; TNFSF1) and TNF (TNFSF2), and CD70 (TNFSF7) and 4-1BBL (TNFSF9), which diverged by tandem duplications early in tetrapod and mammalian evolution, respectively. Although lacking one-to-one LTA and TNF orthologs, sharks have evolved lineage-specific clusters of LTA/TNF co-orthologs. Other key findings include the presence of two BAFF (TNFSF13B) genes along with orthologs of APRIL (TNFSF13) and BALM (TNFSF13C) in sharks, and that all cartilaginous fish genomes harbor an ∼400-million-year-old cluster of multiple FASLG (TNFSF6) orthologs. Finally, sharks have retained seven ancestral jawed vertebrate TNFSF genes lost in humans. Taken together, our data indicate that the jawed vertebrate ancestor possessed a much larger and diverse TNFSF repertoire than previously hypothesized and oppose the idea that the cartilaginous fish immune system is “primitive” compared with that of mammals.
Collapse
Affiliation(s)
- Anthony K. Redmond
- *Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
- †Department of Science and Health, Institute of Technology Carlow, Carlow, Ireland
| | - Rita Pettinello
- ‡School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Fiona K. Bakke
- ‡School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Helen Dooley
- §Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD; and
- ¶Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
3
|
Zhao M, Fu L, Chai Y, Sun M, Li Y, Wang S, Qi J, Zeng B, Kang L, Gao GF, Tan S. Atypical TNF-TNFR superfamily binding interface in the GITR-GITRL complex for T cell activation. Cell Rep 2021; 36:109734. [PMID: 34551288 DOI: 10.1016/j.celrep.2021.109734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/16/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022] Open
Abstract
Glucocorticoid-induced tumor necrosis factor receptor family-related protein (GITR) is a critical regulatory molecule in modulation of T cell immune responses. Here we report the mouse GITR (mGITR) and mGITR ligand (mGITRL) complex structure and find that the binding interface of mGITR and mGITRL is distinct from the typical tumor necrosis factor superfamily (TNFSF)/TNF receptor superfamily (TNFRSF) members. mGITR binds to its ligand with a single domain, whereas the binding interface on mGITRL is located on the side, which is distal from conserved binding sites of TNFSF molecules. Mutational analysis reveals that the binding interface of GITR/GITRL in humans is conserved with that in the mouse. Substitution of key interacting D93-I94-V95 (DIV) in mGITR with the corresponding K93-F94-S95 (KFS) in human GITR enables cross-recognition with human GITRL and cross-activation of receptor signaling. The findings of this study substantially expand our understanding of the interaction of TNFSF/TNFRSF superfamily molecules and can benefit the future design of biologics by targeting GITR/GITRL.
Collapse
Affiliation(s)
- Min Zhao
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Lijun Fu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yan Chai
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Meng Sun
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shuo Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianxun Qi
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bin Zeng
- College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Le Kang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - George F Gao
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shuguang Tan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
4
|
Alvarez-de Miranda FJ, Alonso-Sánchez I, Alcamí A, Hernaez B. TNF Decoy Receptors Encoded by Poxviruses. Pathogens 2021; 10:pathogens10081065. [PMID: 34451529 PMCID: PMC8401223 DOI: 10.3390/pathogens10081065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/29/2021] [Accepted: 08/18/2021] [Indexed: 12/16/2022] Open
Abstract
Tumour necrosis factor (TNF) is an inflammatory cytokine produced in response to viral infections that promotes the recruitment and activation of leukocytes to sites of infection. This TNF-based host response is essential to limit virus spreading, thus poxviruses have evolutionarily adopted diverse molecular mechanisms to counteract TNF antiviral action. These include the expression of poxvirus-encoded soluble receptors or proteins able to bind and neutralize TNF and other members of the TNF ligand superfamily, acting as decoy receptors. This article reviews in detail the various TNF decoy receptors identified to date in the genomes from different poxvirus species, with a special focus on their impact on poxvirus pathogenesis and their potential use as therapeutic molecules.
Collapse
|
5
|
Wang F, Chau B, West SM, Kimberlin CR, Cao F, Schwarz F, Aguilar B, Han M, Morishige W, Bee C, Dollinger G, Rajpal A, Strop P. Structures of mouse and human GITR-GITRL complexes reveal unique TNF superfamily interactions. Nat Commun 2021; 12:1378. [PMID: 33654081 PMCID: PMC7925557 DOI: 10.1038/s41467-021-21563-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/02/2021] [Indexed: 01/10/2023] Open
Abstract
Glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) and GITR ligand (GITRL) are members of the tumor necrosis superfamily that play a role in immune cell signaling, activation, and survival. GITR is a therapeutic target for directly activating effector CD4 and CD8 T cells, or depleting GITR-expressing regulatory T cells (Tregs), thereby promoting anti-tumor immune responses. GITR activation through its native ligand is important for understanding immune signaling, but GITR structure has not been reported. Here we present structures of human and mouse GITR receptors bound to their cognate ligands. Both species share a receptor–ligand interface and receptor–receptor interface; the unique C-terminal receptor–receptor enables higher order structures on the membrane. Human GITR–GITRL has potential to form a hexameric network of membrane complexes, while murine GITR–GITRL complex forms a linear chain due to dimeric interactions. Mutations at the receptor–receptor interface in human GITR reduce cell signaling with in vitro ligand binding assays and minimize higher order membrane structures when bound by fluorescently labeled ligand in cell imaging experiments. Glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) and GITR ligand (GITRL) regulate immune cell activities, including anti-tumor immune responses. Structures and visualization of human and mouse GITR–GITRL complexes offer insight into the architecture of higher-order membrane assemblies, and their signaling.
Collapse
Affiliation(s)
- Feng Wang
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Bryant Chau
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Sean M West
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | | | - Fei Cao
- Discovery Chemistry, Bristol Myers Squibb, Redwood City, CA, USA
| | - Flavio Schwarz
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Barbara Aguilar
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Minhua Han
- Tumor Microenvironment Thematic Research Center, Bristol Myers Squibb, Redwood City, CA, USA
| | - Winse Morishige
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Christine Bee
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Gavin Dollinger
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Arvind Rajpal
- Genentech Research and Early Development, South San Francisco, CA, USA
| | - Pavel Strop
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA.
| |
Collapse
|
6
|
Li Y, Tan S, Zhang C, Chai Y, He M, Zhang CWH, Wang Q, Tong Z, Liu K, Lei Y, Liu WJ, Liu Y, Tian Z, Cao X, Yan J, Qi J, Tien P, Gao S, Gao GF. Limited Cross-Linking of 4-1BB by 4-1BB Ligand and the Agonist Monoclonal Antibody Utomilumab. Cell Rep 2019; 25:909-920.e4. [PMID: 30355497 DOI: 10.1016/j.celrep.2018.09.073] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/13/2018] [Accepted: 09/21/2018] [Indexed: 10/28/2022] Open
Abstract
Monoclonal antibodies (mAbs) targeting the co-stimulatory molecule 4-1BB are of interest for tumor immunotherapy. We determined the complex structures of human 4-1BB with 4-1BB ligand (4-1BBL) or utomilumab to elucidate the structural basis of 4-1BB activation. The 4-1BB/4-1BBL complex displays a typical TNF/TNFR family binding mode. The structure of utomilumab/4-1BB complex shows that utomilumab binds to dimeric 4-1BB with a distinct but partially overlapping binding area with 4-1BBL. Competitive binding analysis demonstrates that utomilumab blocks the 4-1BB/4-1BBL interaction, indicating the interruption of ligand-mediated signaling. The binding profiles of 4-1BBL and utomilumab to monomeric or dimeric 4-1BB indicate limited cross-linking of 4-1BB molecules. These findings provide mechanistic insight into the binding of 4-1BB with its ligand and its agonist mAb, which may facilitate the future development of anti-4-1BB biologics for tumor immunotherapy.
Collapse
Affiliation(s)
- Yan Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuguang Tan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China
| | - Chang Zhang
- CAS Key Laboratory of Bio-medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yan Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Mengnan He
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Qihui Wang
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhou Tong
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China
| | - Kefang Liu
- University of Chinese Academy of Sciences, Beijing 100049, China; National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Yifan Lei
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - William J Liu
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China
| | - Yingxia Liu
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei 230027, China
| | - Xuetao Cao
- Department of Immunology & Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Jinghua Yan
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Po Tien
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Shan Gao
- CAS Key Laboratory of Bio-medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China.
| | - George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen 518112, China; Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China; National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing 102206, China.
| |
Collapse
|
7
|
Das S, Sutoh Y, Cancro MP, Rast JP, Han Q, Bommakanti G, Cooper MD, Hirano M. Ancient BCMA-like Genes Herald B Cell Regulation in Lampreys. THE JOURNAL OF IMMUNOLOGY 2019; 203:2909-2916. [PMID: 31666307 DOI: 10.4049/jimmunol.1900026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 09/25/2019] [Indexed: 11/19/2022]
Abstract
The TNF superfamily ligands BAFF and APRIL interact with three receptors, BAFFR, BCMA, and TACI, to play discrete and crucial roles in regulating B cell selection and homeostasis in mammals. The interactions between these ligands and receptors are both specific and redundant: BAFFR binds BAFF, whereas BCMA and TACI bind to either BAFF or APRIL. In a previous phylogenetic inquiry, we identified and characterized a BAFF-like gene in lampreys, which, with hagfish, are the only extant jawless vertebrates, both of which have B-like and T-like lymphocytes. To gain insight into lymphocyte regulation in jawless vertebrates, in this study we identified two BCMA-like genes in lampreys, BCMAL1 and BCMAL2, which were found to be preferentially expressed by B-like lymphocytes. In vitro analyses indicated that the lamprey BAFF-like protein can bind to a BCMA-like receptor Ig fusion protein and to both BCMAL1- and BCMAL2-transfected cells. Discriminating regulatory roles for the two BCMA-like molecules are suggested by their differential expression before and after activation of the B-like lymphocytes in lampreys. Our composite results imply that BAFF-based mechanisms for B cell regulation evolved before the divergence of jawed and jawless vertebrates.
Collapse
Affiliation(s)
- Sabyasachi Das
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322; .,Emory Vaccine Center, Emory University, Atlanta, GA 30317; and
| | - Yoichi Sutoh
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322.,Emory Vaccine Center, Emory University, Atlanta, GA 30317; and
| | - Michael P Cancro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jonathan P Rast
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322.,Emory Vaccine Center, Emory University, Atlanta, GA 30317; and
| | - Qifeng Han
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322.,Emory Vaccine Center, Emory University, Atlanta, GA 30317; and
| | - Gayathri Bommakanti
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322.,Emory Vaccine Center, Emory University, Atlanta, GA 30317; and
| | - Max D Cooper
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322.,Emory Vaccine Center, Emory University, Atlanta, GA 30317; and
| | - Masayuki Hirano
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322; .,Emory Vaccine Center, Emory University, Atlanta, GA 30317; and
| |
Collapse
|
8
|
Wang YR, Wang J, Liu YH, Hu GL, Gao CY, Wang YJ, Zhou XF, Zeng F. Cysteine-Rich Repeat Domains 2 and 4 are Amyloid-β Binding Domains of Neurotrophin Receptor p75NTR and Potential Targets to Block Amyloid-β Neurotoxicity. J Alzheimers Dis 2019; 63:139-147. [PMID: 29578485 DOI: 10.3233/jad-171012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The p75 neurotrophin receptor (p75NTR) is an amyloid-β (Aβ) receptor that both mediates Aβ neurotoxicity and regulates Aβ production and deposition, thus playing an important role in the pathogenesis of Alzheimer's disease (AD). The extracellular domain of p75NTR (p75ECD), consisting of four cysteine-rich repeat domains (CRDs), was recently reported to be an endogenous anti-Aβ scavenger to block p75NTR-mediated neuronal death and neurite degeneration signaling of Aβ and pro-neurotrophins. Identification of the specific Aβ binding domains of p75NTR is crucial for illuminating their interactions and the etiology of AD. CRDs of p75ECD were obtained by expression of recombinant plasmids or direct synthesis. Aβ aggregation inhibiting test and immunoprecipitation assay were applied to locate the specific binding domains of Aβ to p75ECD. The Aβ neurotoxicity antagonistic effects of different CRDs were examined by cytotoxicity experiments including neurite outgrowth assay, propidium iodide (PI) staining, and MTT assay. In the Aβ aggregation inhibiting test, the fluorescence intensity in the CRD2 and CRD4 treatment groups was significantly lower than that in the CRD1 and CRD3 treatment groups. Immunoprecipitation assay and western blot confirmed that Aβ could bind to CRD2 and CRD4. Besides, CRD2 and CRD4 antagonized Aβ neurotoxicity suggested by longer neurite length, less PI labelled cells, and higher cell viability than the control group. Our results indicate that CRD2 and CRD4 are Aβ binding domains of p75NTR and capable of antagonizing Aβ neurotoxicity, and therefore are potential therapeutic targets to block the interaction of Aβ and p75NTR in the pathogenesis of AD.
Collapse
Affiliation(s)
- Ye-Ran Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital and Research Institute of Field Surgery, Third Military Medical University, Yuzhong district, Chongqing, China
| | - Jun Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital and Research Institute of Field Surgery, Third Military Medical University, Yuzhong district, Chongqing, China
| | - Yu-Hui Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital and Research Institute of Field Surgery, Third Military Medical University, Yuzhong district, Chongqing, China
| | - Gong-Ling Hu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital and Research Institute of Field Surgery, Third Military Medical University, Yuzhong district, Chongqing, China
| | - Chang-Yue Gao
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital and Research Institute of Field Surgery, Third Military Medical University, Yuzhong district, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital and Research Institute of Field Surgery, Third Military Medical University, Yuzhong district, Chongqing, China
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences and Sansom Institute, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Fan Zeng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital and Research Institute of Field Surgery, Third Military Medical University, Yuzhong district, Chongqing, China
| |
Collapse
|
9
|
Zhao Y, Li Y, Qu R, Chen X, Wang W, Qiu C, Liu B, Pan X, Liu L, Vasilev K, Hayball J, Dong S, Li W. Cortistatin binds to TNF-α receptors and protects against osteoarthritis. EBioMedicine 2019; 41:556-570. [PMID: 30826358 PMCID: PMC6443028 DOI: 10.1016/j.ebiom.2019.02.035] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/06/2019] [Accepted: 02/15/2019] [Indexed: 02/06/2023] Open
Abstract
Background Osteoarthritis (OA) is a common degenerative disease, and tumor necrosis factor (TNF-α) is known to play a critical role in OA. Cortistatin (CST) is a neuropeptide discovered over 20 years ago, which plays a vital role in inflammatory reactions. However, it is unknown whether CST is involved in cartilage degeneration and OA development. Methods The interaction between CST and TNF-α receptors was investigated through Coimmunoprecipitation and Biotin-based solid-phase binding assay. Western blot, Real-time PCR, ELISA, immunofluorescence staining, nitrite production assay and DMMB assay of GAG were performed for the primary chondrocyte experiments. Surgically induced and spontaneous OA models were established and western blot, flow cytometry, Real-time PCR, ELISA, immunohistochemistry and fluorescence in vivo imaging were performed for in vivo experiments. Findings CST competitively bound to TNFR1 as well as TNFR2. CST suppressed proinflammatory function of TNF-α. Both spontaneous and surgically induced OA models indicated that deficiency of CST led to an accelerated OA-like phenotype, while exogenous CST attenuated OA development in vivo. Additionally, TNFR1- and TNFR2-knockout mice were used for analysis and indicated that TNFRs might be involved in the protective role of CST in OA. CST inhibited activation of the NF-κB signaling pathway in OA. Interpretation This study provides new insight into the pathogenesis and therapeutic strategy of cartilage degenerative diseases, including OA. Fund The National Natural Science Foundation of China, the Natural Science Foundation of Shandong Province, Key Research and Development Projects of Shandong Province and the Cross-disciplinary Fund of Shandong University.
Collapse
Affiliation(s)
- Yunpeng Zhao
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Yuhua Li
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Ruize Qu
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China; Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012. PR China
| | - Xiaomin Chen
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China; Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012. PR China
| | - Wenhan Wang
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China; Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012. PR China
| | - Cheng Qiu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012. PR China
| | - Ben Liu
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Xin Pan
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China
| | - Liang Liu
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Krasimir Vasilev
- Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia; School of Engineering, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - John Hayball
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Shuli Dong
- College of Chemistry, Shandong University, Jinan, Shandong 250101, PR China
| | - Weiwei Li
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
10
|
Pontejo SM, Sanchez C, Ruiz-Argüello B, Alcami A. Insights into ligand binding by a viral tumor necrosis factor (TNF) decoy receptor yield a selective soluble human type 2 TNF receptor. J Biol Chem 2019; 294:5214-5227. [PMID: 30723161 DOI: 10.1074/jbc.ra118.005828] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/17/2019] [Indexed: 12/20/2022] Open
Abstract
Etanercept is a soluble form of the tumor necrosis factor receptor 2 (TNFR2) that inhibits pathological tumor necrosis factor (TNF) responses in rheumatoid arthritis and other inflammatory diseases. However, besides TNF, etanercept also blocks lymphotoxin-α (LTα), which has no clear therapeutic value and might aggravate some of the adverse effects associated with etanercept. Poxviruses encode soluble TNFR2 homologs, termed viral TNF decoy receptors (vTNFRs), that display unique specificity properties. For instance, cytokine response modifier D (CrmD) inhibits mouse and human TNF and mouse LTα, but it is inactive against human LTα. Here, we analyzed the molecular basis of these immunomodulatory activities in the ectromelia virus-encoded CrmD. We found that the overall molecular mechanism to bind TNF and LTα from mouse and human origin is fairly conserved in CrmD and dominated by a groove under its 50s loop. However, other ligand-specific binding determinants optimize CrmD for the inhibition of mouse ligands, especially mouse TNF. Moreover, we show that the inability of CrmD to inhibit human LTα is caused by a Glu-Phe-Glu motif in its 90s loop. Importantly, transfer of this motif to etanercept diminished its anti-LTα activity in >60-fold while weakening its TNF-inhibitory capacity in 3-fold. This new etanercept variant could potentially be used in the clinic as a safer alternative to conventional etanercept. This work is the most detailed study of the vTNFR-ligand interactions to date and illustrates that a better knowledge of vTNFRs can provide valuable information to improve current anti-TNF therapies.
Collapse
Affiliation(s)
- Sergio M Pontejo
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Carolina Sanchez
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Begoña Ruiz-Argüello
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Antonio Alcami
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
11
|
Zapata JM, Perez-Chacon G, Carr-Baena P, Martinez-Forero I, Azpilikueta A, Otano I, Melero I. CD137 (4-1BB) Signalosome: Complexity Is a Matter of TRAFs. Front Immunol 2018; 9:2618. [PMID: 30524423 PMCID: PMC6262405 DOI: 10.3389/fimmu.2018.02618] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022] Open
Abstract
CD137 (4-1BB, Tnsfr9) is a member of the TNF-receptor (TNFR) superfamily without known intrinsic enzymatic activity in its cytoplasmic domain. Hence, akin to other members of the TNFR family, it relies on the TNFR-Associated-Factor (TRAF) family of adaptor proteins to build the CD137 signalosome for transducing signals into the cell. Thus, upon CD137 activation by binding of CD137L trimers or by crosslinking with agonist monoclonal antibodies, TRAF1, TRAF2, and TRAF3 are readily recruited to the cytoplasmic domain of CD137, likely as homo- and/or heterotrimers with different configurations, initiating the construction of the CD137 signalosome. The formation of TRAF2-RING dimers between TRAF2 molecules from contiguous trimers would help to establish a multimeric structure of TRAF-trimers that is probably essential for CD137 signaling. In addition, available studies have identified a large number of proteins that are recruited to CD137:TRAF complexes including ubiquitin ligases and proteases, kinases, and modulatory proteins. Working in a coordinated fashion, these CD137-signalosomes will ultimately promote CD137-mediated T cell proliferation and survival and will endow T cells with stronger effector functions. Current evidence allows to envision the molecular events that might take place in the early stages of CD137-signalosome formation, underscoring the key roles of TRAFs and of K63 and K48-ubiquitination of target proteins in the signaling process. Understanding the composition and fine regulation of CD137-signalosomes assembly and disassembly will be key to improve the therapeutic activities of chimeric antigen receptors (CARs) encompassing the CD137 cytoplasmic domain and a new generation of CD137 agonists for the treatment of cancer.
Collapse
Affiliation(s)
- Juan M Zapata
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Gema Perez-Chacon
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Pablo Carr-Baena
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Ivan Martinez-Forero
- Departamento de Inmunologia and Inmunoterapia, Centro de Investigación Medica Aplicada, Universidad de Navarra, Pamplona, Spain
| | - Arantza Azpilikueta
- Departamento de Inmunologia and Inmunoterapia, Centro de Investigación Medica Aplicada, Universidad de Navarra, Pamplona, Spain
| | - Itziar Otano
- Departamento de Inmunologia and Inmunoterapia, Centro de Investigación Medica Aplicada, Universidad de Navarra, Pamplona, Spain
| | - Ignacio Melero
- Departamento de Inmunologia and Inmunoterapia, Centro de Investigación Medica Aplicada, Universidad de Navarra, Pamplona, Spain.,MSD, London, United Kingdom.,Departamento de Inmunologia e Inmunoterapia, Clinica Universitaria, Universidad de Navarra, Pamplona, Spain.,Instituto de Investigacion Sanitaria de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
12
|
Jacob JM, Subramaniam K, Tu SL, Nielsen O, Tuomi PA, Upton C, Waltzek TB. Complete genome sequence of a novel sea otterpox virus. Virus Genes 2018; 54:756-767. [PMID: 30225673 DOI: 10.1007/s11262-018-1594-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/07/2018] [Indexed: 11/28/2022]
Abstract
Members of the Poxviridae family are large, double-stranded DNA viruses that replicate in the cytoplasm of their host cells. The subfamily Chordopoxvirinae contains viruses that infect a wide range of vertebrates including marine mammals within the Balaenidae, Delphinidae, Mustelidae, Odobenidae, Otariidae, Phocidae, and Phocoenidae families. Recently, a novel poxvirus was found in a northern sea otter pup (Enhydra lutris kenyoni) that stranded in Alaska in 2009. The phylogenetic relationships of marine mammal poxviruses are not well established because of the lack of complete genome sequences. The current study sequenced the entire sea otterpox virus Enhydra lutris kenyoni (SOPV-ELK) genome using an Illumina MiSeq sequencer. The SOPV-ELK genome is the smallest poxvirus genome known at 127,879 bp, is 68.7% A+T content, is predicted to encode 132 proteins, and has 2546 bp inverted terminal repeats at each end. Genetic and phylogenetic analyses based on the concatenated amino acid sequences of 7 chorodopoxvirus core genes revealed the SOPV-ELK is 52.5-74.1% divergent from other known chordopoxviruses and is most similar to pteropoxvirus from Australia (PTPV-Aus). SOPV-ELK represents a new chordopoxvirus species and may belong to a novel genus. SOPV-ELK encodes eight unique genes. While the function of six predicted genes remains unknown, two genes appear to function as novel immune-modulators. SOPV-ELK-003 appears to encode a novel interleukin-18 binding protein (IL-18 BP), based on limited sequence and structural similarity to other poxviral IL-18 BPs. SOPV-ELK-035 appears to encode a novel tumor necrosis factor receptor-like (TNFR) protein that may be associated with the depression of the host's antiviral response. Additionally, SOPV-ELK-036 encodes a tumor necrosis factor-like apoptosis-inducing ligand (TRAIL) protein that has previously only been found in PTPV-Aus. The SOPV-ELK genome is the first mustelid poxvirus and only the second poxvirus from a marine mammal to be fully sequenced. Sequencing of the SOPV-ELK genome is an important step in unraveling the position of marine mammal poxviruses within the larger Poxviridae phylogenetic tree and provides the necessary sequence to develop molecular tools for future diagnostics and epidemiological studies.
Collapse
Affiliation(s)
- Jessica M Jacob
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Kuttichantran Subramaniam
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Shin-Lin Tu
- Biochemistry and Microbiology Department, University of Victoria, Victoria, BC, V8W 2Y2, Canada
| | - Ole Nielsen
- Department of Fisheries and Oceans Canada, Central and Arctic Region, Winnipeg, MB, R3T 2N6, Canada
| | | | - Chris Upton
- Biochemistry and Microbiology Department, University of Victoria, Victoria, BC, V8W 2Y2, Canada
| | - Thomas B Waltzek
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
13
|
Muller J, Baeyens A, Dustin ML. Tumor Necrosis Factor Receptor Superfamily in T Cell Priming and Effector Function. Adv Immunol 2018; 140:21-57. [PMID: 30366518 DOI: 10.1016/bs.ai.2018.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The tumor necrosis factor receptor superfamily (TNFRSF) and their ligands mediate lymphoid tissue development and homeostasis in addition to key aspects of innate and adaptive immune responses. T cells of the adaptive immune system express a number of TNFRSF members that are used to receive signals at different instructive stages and produce several tumor necrosis factor superfamily (TNFSF) members as effector molecules. There is also one example of a TNFRSF member serving as a ligand for negative regulatory checkpoint receptors. In most cases, the ligands in afferent and efferent phases are membrane proteins and thus the interaction with TNFRSF members must take place in immunological synapses and other modes of cell-cell interaction. A particular feature of the TNFRSF-mediated signaling is the prominent use of linear ubiquitin chains as scaffolds for signaling complexes that activate nuclear factor κ-B and Fos/Jun transcriptional regulators. This review will focus on the signaling mechanisms triggered by TNFRSF members in their role as costimulators of early and late phases of T cell instruction and the delivery mechanism of TNFSF members through the immunological synapses of helper and cytotoxic effector cells.
Collapse
Affiliation(s)
- James Muller
- Skirball Institute of Biomolecular Medicine and Immunology Training Program, New York University School of Medicine, New York, NY, United States
| | - Audrey Baeyens
- Skirball Institute of Biomolecular Medicine and Immunology Training Program, New York University School of Medicine, New York, NY, United States
| | - Michael L Dustin
- Skirball Institute of Biomolecular Medicine and Immunology Training Program, New York University School of Medicine, New York, NY, United States; Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
14
|
Choi BK, Kim SH, Kim YH, Lee DG, Oh HS, Han C, Kim YI, Jeon Y, Lee H, Kwon BS. RELT negatively regulates the early phase of the T-cell response in mice. Eur J Immunol 2018; 48:1739-1749. [PMID: 30138536 DOI: 10.1002/eji.201847633] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/18/2018] [Accepted: 08/20/2018] [Indexed: 12/16/2022]
Abstract
RELT (tumor necrosis factor receptor superfamily member 19-like, TNFRSF19L) is a TNFR superfamily member that is primarily expressed in immune cells and lymphoid tissues, but whose immunological function is not well-defined. Here, we show that RELT is expressed by naive T cells and DCs, and their activation or maturation decreases RELT expression. Using RELT knockout (RELT-/- ) mice, we demonstrate that RELT deficiency selectively promotes the homeostatic proliferation of CD4+ T cells but not CD8+ T cells, and enhances anti-tumor CD8+ T-cell responses. We also demonstrate, using an adoptive transfer model in which RELT is knocked-out in either the transferred transgenic CD8+ T cells or the recipient melanoma-bearing mice, that RELT on multiple immune cells limits the hyper-response of tumor-specific CD8+ T cells. Hyper-responsiveness of RELT-deficient T cells was induced by promoting their proliferation. Taken together, our findings suggest that RELT acts as a negative regulator that controls the early phase of T-cell activation probably by promoting T-cell apoptosis.
Collapse
Affiliation(s)
- Beom K Choi
- Biomedicine Production Branch, National Cancer Center, Korea
| | - Seon-Hee Kim
- Immunotherapeutics Branch, Division of Convergence Technology, National Cancer Center, Korea
| | - Young H Kim
- Biomedicine Production Branch, National Cancer Center, Korea.,Eutilex, Co., Ltd., Seoul, Korea
| | - Don G Lee
- Biomedicine Production Branch, National Cancer Center, Korea
| | - Ho S Oh
- Immunotherapeutics Branch, Division of Convergence Technology, National Cancer Center, Korea.,Eutilex, Co., Ltd., Seoul, Korea
| | - Chungyong Han
- Immunotherapeutics Branch, Division of Convergence Technology, National Cancer Center, Korea
| | - Yu I Kim
- Graduate School of Cancer Science and Policy, National Cancer Center, Korea
| | - Yoon Jeon
- Graduate School of Cancer Science and Policy, National Cancer Center, Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Korea
| | - Byoung S Kwon
- Eutilex, Co., Ltd., Seoul, Korea.,Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
15
|
Fellermeier-Kopf S, Gieseke F, Sahin U, Müller D, Pfizenmaier K, Kontermann RE. Duokines: a novel class of dual-acting co-stimulatory molecules acting in cis or trans. Oncoimmunology 2018; 7:e1471442. [PMID: 30228940 PMCID: PMC6140609 DOI: 10.1080/2162402x.2018.1471442] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 11/23/2022] Open
Abstract
Co-stimulatory signals induced by ligands of the tumor necrosis factor superfamily (TNFSF) play a central role in T cell activation and have emerged as a promising strategy in cancer immunotherapy. Here, we established a novel class of bifunctional co-stimulatory fusion proteins with the aim to boost T cell activation at the level of T cell – antigen-presenting cell (APC) interaction. These novel dual-acting cytokine fusion proteins were created by connecting two different homotrimeric TNFSF ligands to form homotrimeric bifunctional molecules (Duokines) or by connecting single-chain derivatives of two different homotrimeric TNFSF with a single, flexible linker (single-chain Duokines, scDuokines). By linking the TNFSF ligands 4-1BBL, OX40L and CD27L in all possible combinations, cis-acting Duokines were generated that act on the same or adjacent T cells, while combining CD40L with 4-1BBL, OX40L and CD27L resulted in trans-acting Duokines acting simultaneously on APCs and T cells. In vitro, co-stimulation of T cells was seen for cis- and trans-acting Duokines and scDuokines in an antigen-independent as well as antigen-specific setting. Trans-acting molecules furthermore activated B cells, which represent a subclass of APCs. In a pilot experiment using the syngeneic B16-FAP mouse tumor model scDuokines displayed antitumoral activity in vivo in combination with a primary T cell-activating bispecific antibody, evident from reduced number of lung metastasis compared to the antibody-only treated group. Our data show that the bifunctional, co-stimulatory duokines are capable to enhance T cell-mediated anti-tumor immune responses, suggesting that they can serve as a new class of immuno-stimulatory molecules for use in cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Sina Fellermeier-Kopf
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,BioNTech RNA Pharmaceuticals GmbH, Mainz, Germany
| | | | - Ugur Sahin
- BioNTech RNA Pharmaceuticals GmbH, Mainz, Germany
| | - Dafne Müller
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
16
|
Maeda T, Suetake H, Odaka T, Miyadai T. Original Ligand for LTβR Is LIGHT: Insight into Evolution of the LT/LTβR System. THE JOURNAL OF IMMUNOLOGY 2018; 201:202-214. [PMID: 29769272 DOI: 10.4049/jimmunol.1700900] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 04/25/2018] [Indexed: 01/23/2023]
Abstract
The lymphotoxin (LT)/LTβ receptor (LTβR) axis is crucial for the regulation of immune responses and development of lymphoid tissues in mammals. Despite the importance of this pathway, the existence and function of LT and LTβR remain obscure for nonmammalian species. In this study, we report a nonmammalian LTβR and its ligand. We demonstrate that TNF-New (TNFN), which has been considered orthologous to mammalian LT, was expressed on the cell surface as a homomer in vitro. This different protein structure indicates that TNFN is not orthologous to mammalian LTα and LTβ. Additionally, we found that LTβR was conserved in teleosts, but the soluble form of recombinant fugu LTβR did not bind to membrane TNFN under the circumstance tested. Conversely, the LTβR recombinant bound to another ligand, LIGHT, similar to that of mammals. These findings indicate that teleost LTβR is originally a LIGHT receptor. In the cytoplasmic region of fugu LTβR, recombinant fugu LTβR bound to the adaptor protein TNFR-associated factor (TRAF) 2, but little to TRAF3. This difference suggests that teleost LTβR could potentially activate the classical NF-κB pathway with a novel binding domain, but would have little ability to activate an alternative one. Collectively, our results suggested that LIGHT was the original ligand for LTβR, and that the teleost immune system lacked the LT/LTβR pathway. Acquisition of the LT ligand and TRAF binding domain after lobe-finned fish may have facilitated the sophistication of the immune system and lymphoid tissues.
Collapse
Affiliation(s)
- Tomoki Maeda
- Graduate School of Biosciences and Biotechnology, Fukui Prefectural University, Fukui 917-0003, Japan.,Japan Society for the Promotion of Science, Tokyo 102-0083, Japan; and
| | - Hiroaki Suetake
- Faculty of Marine Science and Technology, Fukui Prefectural University, Fukui 917-0003, Japan
| | - Tomoyuki Odaka
- Faculty of Marine Science and Technology, Fukui Prefectural University, Fukui 917-0003, Japan
| | - Toshiaki Miyadai
- Faculty of Marine Science and Technology, Fukui Prefectural University, Fukui 917-0003, Japan
| |
Collapse
|
17
|
Gilbreth RN, Oganesyan VY, Amdouni H, Novarra S, Grinberg L, Barnes A, Baca M. Crystal structure of the human 4-1BB/4-1BBL complex. J Biol Chem 2018; 293:9880-9891. [PMID: 29720399 DOI: 10.1074/jbc.ra118.002803] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Indexed: 11/06/2022] Open
Abstract
4-1BBL is a member of the tumor necrosis factor (TNF) superfamily and is the ligand for the TNFR superfamily receptor, 4-1BB. 4-1BB plays an immunomodulatory role in T cells and NK cells, and agonists of this receptor have garnered strong attention as potential immunotherapy agents. Broadly speaking, the structural features of TNF superfamily members, their receptors, and ligand-receptor complexes are similar. However, a published crystal structure of human 4-1BBL suggests that it may be unique in this regard, exhibiting a three-bladed propeller-like trimer assembly that is distinctly different from that observed in other family members. This unusual structure also suggests that the human 4-1BB/4-1BBL complex may be structurally unique within the TNF/TNFR superfamily, but to date no structural data have been reported. Here we report the crystal structure of the human 4-1BB/4-1BBL complex at 2.4-Å resolution. In this structure, 4-1BBL does not adopt the unusual trimer assembly previously reported, but instead forms a canonical bell-shaped trimer typical of other TNF superfamily members. The structure of 4-1BB is also largely canonical as is the 4-1BB/4-1BBL complex. Mutational data support the 4-1BBL structure reported here as being biologically relevant, suggesting that the previously reported structure is not. Together, the data presented here offer insight into structure/function relationships in the 4-1BB/4-1BBL system and improve our structural understanding of the TNF/TNFR superfamily more broadly.
Collapse
Affiliation(s)
- Ryan N Gilbreth
- From the Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland 20878
| | - Vaheh Y Oganesyan
- From the Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland 20878
| | - Hamza Amdouni
- From the Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland 20878
| | - Shabazz Novarra
- From the Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland 20878
| | - Luba Grinberg
- From the Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland 20878
| | - Arnita Barnes
- From the Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland 20878
| | - Manuel Baca
- From the Department of Antibody Discovery and Protein Engineering, MedImmune LLC, Gaithersburg, Maryland 20878
| |
Collapse
|
18
|
Zhou J, Wu HG, Shi Y. Roles of TNF-α/NF-κB/Snail pathway in regulating epithelial-mesenchymal transition. Shijie Huaren Xiaohua Zazhi 2018; 26:441-448. [DOI: 10.11569/wcjd.v26.i7.441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process of transformation of epithelial cells to mesenchymal cells, and it not only plays an important role in the developmental process, but also participates in tissue healing, organ fibrosis, tumorigenesis, and metastasis. In recent years, it has been found that tumor necrosis factor-α (TNF-α) is a major inflammatory factor that can induce snail expression by binding to nuclear factor-κB (NF-κB), thus mediating EMT. This article briefly introduces the roles of the TNF-α/NF-κB/Snail pathway in mediating EMT, aiming to promote a further understanding of the mechanism of TNF-α in regulating EMT.
Collapse
|
19
|
Bitra A, Doukov T, Wang J, Picarda G, Benedict CA, Croft M, Zajonc DM. Crystal structure of murine 4-1BB and its interaction with 4-1BBL support a role for galectin-9 in 4-1BB signaling. J Biol Chem 2018; 293:1317-1329. [PMID: 29242193 PMCID: PMC5787808 DOI: 10.1074/jbc.m117.814905] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/01/2017] [Indexed: 11/06/2022] Open
Abstract
4-1BB (CD137) is a TNF receptor superfamily (TNFRSF) member that is thought to undergo receptor trimerization upon binding to its trimeric TNF superfamily ligand (4-1BBL) to stimulate immune responses. 4-1BB also can bind to the tandem repeat-type lectin galectin-9 (Gal-9), and signaling through mouse (m)4-1BB is reduced in galectin-9 (Gal-9)-deficient mice, suggesting a pivotal role of Gal-9 in m4-1BB activation. Here, using sulfur-SAD phasing, we determined the crystal structure of m4-1BB to 2.2-Å resolution. We found that similar to other TNFRSFs, m4-1BB has four cysteine-rich domains (CRDs). However, the organization of CRD1 and the orientation of CRD3 and CRD4 with respect to CRD2 in the m4-1BB structure distinctly differed from those of other TNFRSFs. Moreover, we mapped two Asn residues within CRD4 that are N-linked glycosylated and mediate m4-1BB binding to Gal-9. Kinetics studies of m4-1BB disclosed a very tight nanomolar binding affinity to m4-1BBL with an unexpectedly strong avidity effect. Both N- and C-terminal domains of Gal-9 bound m4-1BB, but with lower affinity compared with m4-1BBL. Although the TNF homology domain (THD) of human (h)4-1BBL forms non-covalent trimers, we found that m4-1BBL formed a covalent dimer via 2 cysteines absent in h4-1BBL. As multimerization and clustering is a prerequisite for TNFR intracellular signaling, and as m4-1BBL can only recruit two m4-1BB monomers, we hypothesize that m4-1BBL and Gal-9 act together to aid aggregation of m4-1BB monomers to efficiently initiate m4-1BB signaling.
Collapse
Affiliation(s)
- Aruna Bitra
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Tzanko Doukov
- the Stanford Synchrotron Radiation Light Source, Menlo Park, California 94025
| | - Jing Wang
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Gaelle Picarda
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Chris A Benedict
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Michael Croft
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
- the Department of Medicine, University of California San Diego, La Jolla, California 92037, and
| | - Dirk M Zajonc
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
- the Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
20
|
Characterization of BAFF and APRIL subfamily receptors in rainbow trout (Oncorhynchus mykiss). Potential role of the BAFF / APRIL axis in the pathogenesis of proliferative kidney disease. PLoS One 2017; 12:e0174249. [PMID: 28323891 PMCID: PMC5360319 DOI: 10.1371/journal.pone.0174249] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/06/2017] [Indexed: 12/31/2022] Open
Abstract
Proliferative kidney disease (PKD) is a parasitic infection of salmonid fish characterized by hyper-secretion of immunoglobulins in response to the presence of the myxozoan parasite, Tetracapsuloides bryosalmonae. In this context, we hypothesized that the BAFF/APRIL axis, known to play a major role in B cell differentiation and survival in mammals, could be affected by the parasite and consequently be involved in the apparent shift in normal B cell activity. To regulate B cell activity, BAFF and APRIL bind to transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) and B cell maturation antigen (BCMA), whereas BAFF also binds to BAFF receptor (BAFF-R). In teleost fish, although some BAFF and APRIL sequences have been reported, their receptors have not been identified. Thus, as a first step in the current work, we have identified homologues to mammalian TACI, BCMA and BAFF-R in rainbow trout (Oncorhynchus mykiss), that constitute the first report of BAFF and APRIL receptor sequences in fish. Subsequently we studied the transcriptional modulation of BAFF, APRIL, and the fish-specific related cytokine, BALM and their putative receptors in fish naturally exposed to T. bryosalmonae. Finally, to gain further insights on the functional role that these cytokines play during the course of PKD, we have studied their effect on the survival of kidney IgM+ B cells and on immunoglobulin transcription. Our results support the premise that the BAFF / APRIL axis could play an important role during PKD, which may open the possibility of new therapeutic treatments against the disease.
Collapse
|
21
|
CD137-CRDI is not necessary in the role of contacting its natural ligand. Immunol Cell Biol 2016; 95:24-32. [PMID: 27430526 DOI: 10.1038/icb.2016.64] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 07/07/2016] [Accepted: 07/10/2016] [Indexed: 01/05/2023]
Abstract
Immune checkpoint inhibitors result in impressive clinical responses and are expanding to treat a wide variety of tumors. One common problem is low responses from current clinical trials that only benefit a fraction of patients. One key promising direction is combination therapy to increase clinical benefit. CD137, a well-defined antitumor target, can cause strong co-stimulating activity and break immune tolerance. In this study, the role of CD137-CRDI (cysteine rich domain I) in the binding of CD137-CD137L was further investigated based on our previous work. The results revealed that CRDI-mediated limited CD137 assembly without relying on CD137L. Furthermore, CRDI was not involved in direct contact with CD137L in either mice or humans. Isolated mouse CRDII and human CRDII+CRDIII were proven to be the minimum unit for interface with their respective ligands. Fine-tuning of this signaling may improve CD137-targeting strategy.
Collapse
|
22
|
Affiliation(s)
- Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
23
|
Liu C, Li XX, Gao W, Liu W, Liu DS. Progranulin-derived Atsttrin directly binds to TNFRSF25 (DR3) and inhibits TNF-like ligand 1A (TL1A) activity. PLoS One 2014; 9:e92743. [PMID: 24651300 PMCID: PMC3961393 DOI: 10.1371/journal.pone.0092743] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 02/25/2014] [Indexed: 12/30/2022] Open
Abstract
Atsttrin, a progranulin (PGRN)-derived molecule composed of three TNFR-binding domains of PGRN, binds to TNF receptors (TNFR) and is therapeutic against inflammatory arthritis. Here we screened the associations of Atsttrin and other members in TNFR subfamily, which led to the discovery of TNFRSF25 (DR3) as an additional Atsttrin-interacting member in TNFR family. Similar to TNFR1 and TNFR2, DR3 also directly bound to Atsttrin. The first three cysteine-rich domains (CRD) in the extracellular portion of DR3 were required for this interaction. Atsttrin inhibited the interaction between DR3 and its TNF-Like Ligand 1A (TL1A). In addition, Atsttrin inhibited TL1A-stimulated target gene expressions and neutralized TL1A-enhanced osteoclastogenesis in vitro. Furthermore, Atsttrin ameliorated the pathology in dextran sulfate sodium induced colitis. Taken together, these findings not only provide the new insights into Atsttrin's therapeutic action in inflammatory arthritis, but may also present Atsttrin as a novel biological agent for treating various types of diseases associated with TL1A/DR3 pathway.
Collapse
MESH Headings
- Animals
- Body Weight/drug effects
- Cell Line
- Colitis/chemically induced
- Colitis/metabolism
- Colitis/pathology
- Cysteine/chemistry
- Dextran Sulfate/adverse effects
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Hemorrhage/prevention & control
- Macrophages/metabolism
- Mice
- Multigene Family
- Protein Binding/drug effects
- Protein Interaction Domains and Motifs
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Member 25/chemistry
- Receptors, Tumor Necrosis Factor, Member 25/genetics
- Receptors, Tumor Necrosis Factor, Member 25/metabolism
- Recombinant Fusion Proteins/metabolism
- Recombinant Fusion Proteins/pharmacology
- Tumor Necrosis Factor Ligand Superfamily Member 15/antagonists & inhibitors
- Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism
Collapse
Affiliation(s)
- Cui Liu
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xing-Xia Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Gao
- Department of Nursing, Qilu Hospital of Shandong University, Jinan, China
| | - Wen Liu
- Department of Biotechnology, Taishan Medical University, Taian, China
| | - De-Shan Liu
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, China
- * E-mail:
| |
Collapse
|
24
|
Yi L, Zhao Y, Wang X, Dai M, Hellström KE, Hellström I, Zhang H. Human and mouse CD137 have predominantly different binding CRDs to their respective ligands. PLoS One 2014; 9:e86337. [PMID: 24466035 PMCID: PMC3897701 DOI: 10.1371/journal.pone.0086337] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/06/2013] [Indexed: 11/18/2022] Open
Abstract
Monoclonal antibodies (mAbs) to CD137 (a.k.a. 4-1BB) have anti-tumor efficacy in several animal models and have entered clinical trials in patients with advanced cancer. Importantly, anti-CD137 mAbs can also ameliorate autoimmunity in preclinical models. As an approach to better understand the action of agonistic and antagonistic anti-CD137 mAbs we have mapped the binding region of the CD137 ligand (CD137L) to human and mouse CD137. By investigating the binding of CD137L to cysteine rich domain II (CRDII )and CRDIII of CD137, we found that the binding interface was limited and differed between the two species in that mouse CD137L mainly combined with CRDII and human CD137L mainly combined with CRDIII.
Collapse
Affiliation(s)
- Ling Yi
- Department of Central Laboratory, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
| | - Yanlin Zhao
- Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Xiaojue Wang
- Department of Central Laboratory, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
| | - Min Dai
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Karl Erik Hellström
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Ingegerd Hellström
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Hongtao Zhang
- Department of Central Laboratory, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
25
|
Progranulin directly binds to the CRD2 and CRD3 of TNFR extracellular domains. FEBS Lett 2013; 587:3428-36. [PMID: 24070898 DOI: 10.1016/j.febslet.2013.09.024] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 09/03/2013] [Accepted: 09/10/2013] [Indexed: 11/20/2022]
Abstract
We previously reported that PGRN directly bound to TNF receptors (TNFR) in vitro and in chondrocytes (Tang, et al., Science, 2011). Here we report that PGRN also associated with TNFR in splenocytes, and inhibited the binding of TNFα to immune cells. Proper folding of PGRN is essential for its binding to TNFR, as DTT treatment abolished its binding to TNFR. In contrast, the binding of PGRN to Sortilin was enhanced by DTT. Protein interaction assays with mutants of the TNFR extracellular domain demonstrated that CRD2 and CRD3 of TNFR are important for the interaction with PGRN, similar to the binding to TNFα. Taken together, these findings provide the molecular basis underlying PGRN/TNFR interaction and PGRN-mediated anti-inflammatory activity in various autoimmune diseases and conditions.
Collapse
|
26
|
Magis C, Di Tommaso P, Notredame C. T-RMSD: a web server for automated fine-grained protein structural classification. Nucleic Acids Res 2013; 41:W358-62. [PMID: 23716642 PMCID: PMC3692075 DOI: 10.1093/nar/gkt383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This article introduces the T-RMSD web server (tree-based on root-mean-square deviation), a service allowing the online computation of structure-based protein classification. It has been developed to address the relation between structural and functional similarity in proteins, and it allows a fine-grained structural clustering of a given protein family or group of structurally related proteins using distance RMSD (dRMSD) variations. These distances are computed between all pairs of equivalent residues, as defined by the ungapped columns within a given multiple sequence alignment. Using these generated distance matrices (one per equivalent position), T-RMSD produces a structural tree with support values for each cluster node, reminiscent of bootstrap values. These values, associated with the tree topology, allow a quantitative estimate of structural distances between proteins or group of proteins defined by the tree topology. The clusters thus defined have been shown to be structurally and functionally informative. The T-RMSD web server is a free website open to all users and available at http://tcoffee.crg.cat/apps/tcoffee/do:trmsd.
Collapse
Affiliation(s)
- Cedrik Magis
- Bioinformatics and Genomics Programme, Centre For Genomic Regulation, Carrer del Doctor Aiguader 88, 08003 Barcelona, Spain
| | | | | |
Collapse
|
27
|
Abstract
Co-stimulatory and co-inhibitory receptors have a pivotal role in T cell biology, as they determine the functional outcome of T cell receptor (TCR) signalling. The classic definition of T cell co-stimulation continues to evolve through the identification of new co-stimulatory and co-inhibitory receptors, the biochemical characterization of their downstream signalling events and the delineation of their immunological functions. Notably, it has been recently appreciated that co-stimulatory and co-inhibitory receptors display great diversity in expression, structure and function, and that their functions are largely context dependent. Here, we focus on some of these emerging concepts and review the mechanisms through which T cell activation, differentiation and function is controlled by co-stimulatory and co-inhibitory receptors.
Collapse
Affiliation(s)
- Lieping Chen
- Department of Immunobiology and Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06519, USA.
| | | |
Collapse
|
28
|
Protein-protein interactions and multi-component complexes of aminoacyl-tRNA synthetases. Top Curr Chem (Cham) 2013; 344:119-44. [PMID: 24072587 DOI: 10.1007/128_2013_479] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Protein-protein interaction occurs transiently or stably when two or more proteins bind together to mediate a wide range of cellular processes such as protein modification, signal transduction, protein trafficking, and structural folding. The macromolecules involved in protein biosynthesis such as aminoacyl-tRNA synthetase (ARS) have a number of protein-protein interactions. The mammalian multi-tRNA synthetase complex (MSC) consists of eight different enzymes: EPRS, IRS, LRS, QRS, MRS, KRS, RRS, and DRS, and three auxiliary proteins: AIMP1/p43, AIMP2/p38, and AIMP/p18. The distinct ARS proteins are also connected to diverse protein networks to carry out biological functions. In this chapter we first show the protein networks of the entire MSC and explain how MSC components interact with or can regulate other proteins. Finally, it is pointed out that the understanding of protein-protein interaction mechanism will provide insight to potential therapeutic application for diseases related to the MSC network.
Collapse
|