1
|
Yu Y, Ba X, Li T, Xu W, Zhao J, Zhang N, Zhao Y, Wang T, Zhang X, Wang X, Bai B, Wang B. PTPN22 and the pathogenesis of ulcerative colitis: Insights into T cell differentiation and the JAK/STAT signaling pathway. Cell Signal 2025; 127:111551. [PMID: 39643025 DOI: 10.1016/j.cellsig.2024.111551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/20/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
70 % of the ulcerative colitis (UC) linked gene loci are associated with other autoimmune or immunodeficient diseases. The phosphatase activity of PTPN22 can regulate the development of T cells and contribute to regulate the level of inflammation in autoimmune diseases. We produced PTPN22-CS thymus-specific transgenic mice, which suppressed PTPN22 enzyme activity in the thymocytes. Overexpressed PTPN22-CS facilitated the development of the thymocytes towards CD4+T cells and resulted in an increased proportion of the Th1 and Treg cells in the UC mesenteric lymph nodes. PTPN22-CS promoted the activation of the JAK/STAT signaling pathway in the Th1 and Treg cells that localized in the colon, resulting in an excessive production of inflammatory mediators such as IL-2 and IFN-γ. Consequently, PTPN22-CS contributes to the inflammatory response of ulcerative colitis. In summary, the tyrosine phosphatase activity of PTPN22 plays a role in modulating UC by regulating T cell differentiation and modulating the JAK/STAT signaling pathway, thereby influencing the inflammatory response in colonic. These findings provide new insight into the association between PTPN22 and the pathogenesis of UC.
Collapse
Affiliation(s)
- Yang Yu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Xinlei Ba
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Tong Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Wenying Xu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Jiahui Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China
| | - Na Zhang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Yanjiao Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China
| | - Tao Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Xiaonan Zhang
- Department of Pathophysiology, Bengbu Medical University, Longzihu, Bengbu, 233030, Anhui, PR China..
| | - Xipeng Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China
| | - Bin Bai
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| | - Bing Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life Science and Health, Northeastern University, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China; College of Life and Health Sciences, Northeastern University, Shenyang, #195 Chuangxin Road, Hunnan Xinqu, Shenyang, Liaoning 110169, China.
| |
Collapse
|
2
|
Patton JT, Woyach JA. Targeting the B cell receptor signaling pathway in chronic lymphocytic leukemia. Semin Hematol 2024; 61:100-108. [PMID: 38749798 DOI: 10.1053/j.seminhematol.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/04/2024] [Accepted: 04/10/2024] [Indexed: 06/09/2024]
Abstract
Aberrant signal transduction through the B cell receptor (BCR) plays a critical role in the pathogenesis of chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL). BCR-dependent signaling is necessary for the growth and survival of neoplastic cells, making inhibition of down-stream pathways a logical therapeutic strategy. Indeed, selective inhibitors against Bruton's tyrosine kinase (BTK) and phosphoinositide 3-kinase (PI3K) have been shown to induce high rates of response in CLL and other B cell lymphomas. In particular, the development of BTK inhibitors revolutionized the treatment approach to CLL, demonstrating long-term efficacy. While BTK inhibitors are widely used for multiple lines of treatment, PI3K inhibitors are much less commonly utilized, mainly due to toxicities. CLL remains an incurable disease and effective treatment options after relapse or development of TKI resistance are greatly needed. This review provides an overview of BCR signaling, a summary of the current therapeutic landscape, and a discussion of the ongoing trials targeting BCR-associated kinases.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Signal Transduction/drug effects
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/antagonists & inhibitors
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors
- Agammaglobulinaemia Tyrosine Kinase/metabolism
- Molecular Targeted Therapy
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
- Phosphoinositide-3 Kinase Inhibitors/therapeutic use
- Phosphoinositide-3 Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- John T Patton
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Jennifer A Woyach
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH.
| |
Collapse
|
3
|
Schlicher L, Green LG, Romagnani A, Renner F. Small molecule inhibitors for cancer immunotherapy and associated biomarkers - the current status. Front Immunol 2023; 14:1297175. [PMID: 38022587 PMCID: PMC10644399 DOI: 10.3389/fimmu.2023.1297175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Following the success of cancer immunotherapy using large molecules against immune checkpoint inhibitors, the concept of using small molecules to interfere with intracellular negative regulators of anti-tumor immune responses has emerged in recent years. The main targets for small molecule drugs currently include enzymes of negative feedback loops in signaling pathways of immune cells and proteins that promote immunosuppressive signals within the tumor microenvironment. In the adaptive immune system, negative regulators of T cell receptor signaling (MAP4K1, DGKα/ζ, CBL-B, PTPN2, PTPN22, SHP1), co-receptor signaling (CBL-B) and cytokine signaling (PTPN2) have been preclinically validated as promising targets and initial clinical trials with small molecule inhibitors are underway. To enhance innate anti-tumor immune responses, inhibitory immunomodulation of cGAS/STING has been in the focus, and inhibitors of ENPP1 and TREX1 have reached the clinic. In addition, immunosuppressive signals via adenosine can be counteracted by CD39 and CD73 inhibition, while suppression via intratumoral immunosuppressive prostaglandin E can be targeted by EP2/EP4 antagonists. Here, we present the status of the most promising small molecule drug candidates for cancer immunotherapy, all residing relatively early in development, and the potential of relevant biomarkers.
Collapse
Affiliation(s)
- Lisa Schlicher
- Cancer Cell Targeted Therapy, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Luke G. Green
- Therapeutic Modalities, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Andrea Romagnani
- Cancer Cell Targeted Therapy, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Florian Renner
- Cancer Cell Targeted Therapy, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| |
Collapse
|
4
|
Zhu S, Wang H, Ranjan K, Zhang D. Regulation, targets and functions of CSK. Front Cell Dev Biol 2023; 11:1206539. [PMID: 37397251 PMCID: PMC10312003 DOI: 10.3389/fcell.2023.1206539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 06/07/2023] [Indexed: 07/04/2023] Open
Abstract
The Src family kinases (SFK) plays an important role in multiple signal transduction pathways. Aberrant activation of SFKs leads to diseases such as cancer, blood disorders, and bone pathologies. By phosphorylating and inactivating SFKs, the C-terminal Src kinase (CSK) serves as the key negative regulator of SFKs. Similar to Src, CSK is composed of SH3, SH2, and a catalytic kinase domain. However, while the Src kinase domain is intrinsically active, the CSK kinase domain is intrinsically inactive. Multiple lines of evidence indicate that CSK is involved in various physiological processes including DNA repair, permeability of intestinal epithelial cells (IECs), synaptic activity, astrocyte-to-neuron communication, erythropoiesis, platelet homeostasis, mast cell activation, immune and inflammation responses. As a result, dysregulation of CSK may lead to many diseases with different underlying molecular mechanisms. Furthermore, recent findings suggest that in addition to the well-established CSK-SFK axis, novel CSK-related targets and modes of CSK regulation also exist. This review focuses on the recent progress in this field for an up-to-date understanding of CSK.
Collapse
Affiliation(s)
- Shudong Zhu
- School of Medicine, Nantong University, Nantong, China
| | - Hui Wang
- School of Medicine, Nantong University, Nantong, China
| | - Kamakshi Ranjan
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| |
Collapse
|
5
|
Stanford SM, Bottini N. Targeting protein phosphatases in cancer immunotherapy and autoimmune disorders. Nat Rev Drug Discov 2023; 22:273-294. [PMID: 36693907 PMCID: PMC9872771 DOI: 10.1038/s41573-022-00618-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 01/25/2023]
Abstract
Protein phosphatases act as key regulators of multiple important cellular processes and are attractive therapeutic targets for various diseases. Although extensive effort has been dedicated to phosphatase-targeted drug discovery, early expeditions for competitive phosphatase inhibitors were plagued by druggability issues, leading to the stigmatization of phosphatases as difficult targets. Despite challenges, persistent efforts have led to the identification of several drug-like, non-competitive modulators of some of these enzymes - including SH2 domain-containing protein tyrosine phosphatase 2, protein tyrosine phosphatase 1B, vascular endothelial protein tyrosine phosphatase and protein phosphatase 1 - reigniting interest in therapeutic targeting of phosphatases. Here, we discuss recent progress in phosphatase drug discovery, with emphasis on the development of selective modulators that exhibit biological activity. The roles and regulation of protein phosphatases in immune cells and their potential as powerful targets for immuno-oncology and autoimmunity indications are assessed.
Collapse
Affiliation(s)
| | - Nunzio Bottini
- Department of Medicine, University of California, San Diego, CA, USA.
| |
Collapse
|
6
|
Sun G, Ayrapetov MK. Dissection of the catalytic and regulatory structure-function relationships of Csk protein tyrosine kinase. Front Cell Dev Biol 2023; 11:1148352. [PMID: 36936693 PMCID: PMC10016382 DOI: 10.3389/fcell.2023.1148352] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
Protein tyrosine kinases (PTKs) are a large enzyme family that regulates many cellular processes. The key to their broad role in signaling is their tunable substrate specificity and regulatory mechanisms that allow each to respond to appropriate regulatory signals and phosphorylate the correct physiological protein substrates. Thus, in addition to the general PTK catalytic platform, each PTK acquires unique structural motifs that confer a unique combination of catalytic and regulatory properties. Understanding the structural basis for these properties is essential for understanding and manipulating the PTK-based signaling networks in normal and cancer cells. C-terminal Src kinase (Csk) and its homolog, Csk-homologous kinase (Chk), phosphorylate Src family kinases on a C-terminal Tyr residue and negatively regulate their kinase activity. While this regulatory function is biologically essential, Csk and Chk have also been excellent model PTKs for dissecting the structural basis of PTK catalysis and regulation. In this article, we review the structure-function studies of Csk and Chk that shed light on the regulatory and catalytic mechanisms of protein tyrosine kinases in general.
Collapse
|
7
|
Protein tyrosine phosphatase PTPN22 negatively modulates platelet function and thrombus formation. Blood 2022; 140:1038-1051. [PMID: 35767715 DOI: 10.1182/blood.2022015554] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/21/2022] [Indexed: 11/20/2022] Open
Abstract
Protein tyrosine phosphatase nonreceptor type 22 (PTPN22) is a protein tyrosine phosphatase that negatively regulates T-cell signaling. However, whether it is expressed and functions in platelets remains unknown. Here we investigated the expression and role of PTPN22 in platelet function. We reported PTPN22 expression in both human and mouse platelets. Using PTPN22-/- mice, we showed that PTPN22 deficiency significantly shortened tail-bleeding time and accelerated arterial thrombus formation without affecting venous thrombosis and the coagulation factors VIII and IX. Consistently, PTPN22-deficient platelets exhibited enhanced platelet aggregation, granule secretion, calcium mobilization, lamellipodia formation, spreading, and clot retraction. Quantitative phosphoproteomic analysis revealed the significant difference of phosphodiesterase 5A (PDE5A) phosphorylation in PTPN22-deficient platelets compared with wild-type platelets after collagen-related peptide stimulation, which was confirmed by increased PDE5A phosphorylation (Ser92) in collagen-related peptide-treated PTPN22-deficient platelets, concomitant with reduced level and vasodilator-stimulated phosphoprotein phosphorylation (Ser157/239). In addition, PTPN22 interacted with phosphorylated PDE5A (Ser92) and dephosphorylated it in activated platelets. Moreover, purified PTPN22 but not the mutant form (C227S) possesses intrinsic serine phosphatase activity. Furthermore, inhibition of PTPN22 enhanced human platelet aggregation, spreading, clot retraction, and increased PDE5A phosphorylation (Ser92). In conclusion, our study shows a novel role of PTPN22 in platelet function and arterial thrombosis, identifying new potential targets for future prevention of thrombotic or cardiovascular diseases.
Collapse
|
8
|
Brian BF, Sjaastad FV, Freedman TS. SH3-domain mutations selectively disrupt Csk homodimerization or PTPN22 binding. Sci Rep 2022; 12:5875. [PMID: 35393453 PMCID: PMC8989918 DOI: 10.1038/s41598-022-09589-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/25/2022] [Indexed: 12/16/2022] Open
Abstract
The kinase Csk is the primary negative regulator of the Src-family kinases (SFKs, e.g., Lck, Fyn, Lyn, Hck, Fgr, Blk, Yes), phosphorylating a tyrosine on the SFK C-terminal tail that mediates autoinhibition. Csk also binds phosphatases, including PTPN12 (PTP-PEST) and immune-cell PTPN22 (LYP/Pep), which dephosphorylate the SFK activation loop to promote autoinhibition. Csk-binding proteins (e.g., CBP/PAG1) oligomerize within membrane microdomains, and high local concentration promotes Csk function. Purified Csk homodimerizes in solution through an interface that overlaps the phosphatase binding footprint. Here we demonstrate that Csk can homodimerize in Jurkat T cells, in competition with PTPN22 binding. We designed SH3-domain mutations in Csk that selectively impair homodimerization (H21I) or PTPN22 binding (K43D) and verified their kinase activity in solution. Disruption of either interaction in cells, however, decreased the negative-regulatory function of Csk. Csk W47A, a substitution previously reported to block PTPN22 binding, had a secondary effect of impairing homodimerization. Csk H21I and K43D will be useful tools for dissecting the protein-specific drivers of autoimmunity mediated by the human polymorphism PTPN22 R620W, which impairs interaction with Csk and with the E3 ubiquitin ligase TRAF3. Future investigations of Csk homodimer activity and phosphatase interactions may reveal new facets of SFK regulation in hematopoietic and non-hematopoietic cells.
Collapse
Affiliation(s)
- Ben F Brian
- Graduate Program in Molecular Pharmacology and Therapeutics, University of Minnesota, Minneapolis, MN, 55455, USA.,Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94170, USA
| | - Frances V Sjaastad
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA.,Department of Cardiac Rhythm Management, Medtronic, Mounds View, MN, 55112, USA
| | - Tanya S Freedman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA. .,Center for Immunology, University of Minnesota, Minneapolis, MN, 55455, USA. .,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA. .,Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
9
|
Orozco RC, Marquardt K, Mowen K, Sherman LA. Proautoimmune Allele of Tyrosine Phosphatase, PTPN22, Enhances Tumor Immunity. THE JOURNAL OF IMMUNOLOGY 2021; 207:1662-1671. [PMID: 34417261 DOI: 10.4049/jimmunol.2100304] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/07/2021] [Indexed: 12/13/2022]
Abstract
The 1858C>T allele of the tyrosine phosphatase PTPN22 (causing amino acid substitution R620W in encoded protein lymphoid tyrosine phosphatase) is present in 5-10% of the North American population and is strongly associated with numerous autoimmune diseases. Although much research has been done to define how this allele potentiates autoimmunity, the influence PTPN22 and its proautoimmune allele have in tumor immunity is poorly defined. To interrogate the role this allele may have in the antitumor immune response, we used CRISPR/Cas9 to generate mice in which the ortholog of lymphoid tyrosine phosphatase, PEST domain-enriched protein (PEP), is mutated at position 619 to produce the relevant proautoimmune mutation (R619W). Results of this study show that mice homozygous for this alteration (PEP-619WW) resist tumor growth as compared with wild-type mice. Consistent with these results, tumors from PEP-619WW mice have more CD45 infiltrates containing more activated CD8 T cells and CD4 T cells. In addition, there are more conventional dendritic cell type 1 (cDC1) cells and fewer myeloid-derived suppressor cells in tumors from PEP-619WW mice. Interestingly, the tumor-infiltrating PEP-619WW cDC1 cells have decreased PD-L1 expression compared with cDC1 cells from PEP-wild-type mice. Taken together, our data show that the proautoimmune allele of Ptpn22 drives a strong antitumor response in innate and adaptive immune cells resulting in superior control of tumors.
Collapse
Affiliation(s)
- Robin C Orozco
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA
| | - Kristi Marquardt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA
| | - Kerri Mowen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA
| | - Linda A Sherman
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
10
|
Tizaoui K, Terrazzino S, Cargnin S, Lee KH, Gauckler P, Li H, Shin JI, Kronbichler A. The role of PTPN22 in the pathogenesis of autoimmune diseases: A comprehensive review. Semin Arthritis Rheum 2021; 51:513-522. [PMID: 33866147 DOI: 10.1016/j.semarthrit.2021.03.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/16/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
The incidence of autoimmune diseases is increasing worldwide, thus stimulating studies on their etiopathogenesis, derived from a complex interaction between genetic and environmental factors. Genetic association studies have shown the PTPN22 gene as a shared genetic risk factor with implications in multiple autoimmune disorders. By encoding a protein tyrosine phosphatase expressed by the majority of cells belonging to the innate and adaptive immune systems, the PTPN22 gene may have a fundamental role in the development of immune dysfunction. PTPN22 polymorphisms are associated with rheumatoid arthritis, type 1 diabetes, systemic lupus erythematosus, and many other autoimmune conditions. In this review, we discuss the progress in our understanding of how PTPN22 impacts autoimmunity in both humans and animal models. In addition, we highlight the pathogenic significance of the PTPN22 gene, with particular emphasis on its role in T and B cells, and its function in innate immune cells, such as monocytes, dendritic and natural killer cells. We focus particularly on the complexity of PTPN22 interplay with biological processes of the immune system. Findings highlight the importance of studying the function of disease-associated PTPN22 variants in different cell types and open new avenues of investigation with the potential to drive further insights into mechanisms of PTPN22. These new insights will reveal important clues to the molecular mechanisms of prevalent autoimmune diseases and propose new potential therapeutic targets.
Collapse
Affiliation(s)
- Kalthoum Tizaoui
- Department of Basic Sciences, Division of Histology and Immunology, Faculty of Medicine Tunis, Tunis El Manar University, Tunis 1068, Tunisia
| | - Salvatore Terrazzino
- Department of Pharmaceutical Sciences and Interdepartmental Research Center of Pharmacogenetics and Pharmacogenomics (CRIFF), University of Piemonte Orientale, Novara, Italy
| | - Sarah Cargnin
- Department of Pharmaceutical Sciences and Interdepartmental Research Center of Pharmacogenetics and Pharmacogenomics (CRIFF), University of Piemonte Orientale, Novara, Italy
| | - Keum Hwa Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Philipp Gauckler
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | - Han Li
- University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Andreas Kronbichler
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Behavior control of membrane-less protein liquid condensates with metal ion-induced phase separation. Nat Commun 2020; 11:5554. [PMID: 33144560 PMCID: PMC7642319 DOI: 10.1038/s41467-020-19391-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
Phase separation of specific biomolecules into liquid droplet-like condensates is a key mechanism to form membrane-less organelles, which spatio-temporally organize diverse biochemical processes in cells. To investigate the working principles of these biomolecular condensates as dynamic reaction centers, precise control of diverse condensate properties is essential. Here, we design a strategy for metal ion-induced clustering of minimal protein modules to produce liquid protein condensates, the properties of which can be widely varied by simple manipulation of the protein clustering systems. The droplet forming-minimal module contains only a single receptor protein and a binding ligand peptide with a hexahistidine tag for divalent metal ion-mediated clustering. A wide range of protein condensate properties such as droplet forming tendency, droplet morphology, inside protein diffusivity, protein recruitment, and droplet density can be varied by adjusting the nature of receptor/ligand pairs or used metal ions, metal/protein ratios, incubation time, binding motif variation on recruited proteins, and even spacing between receptor/ligand pairs and the hexahistidine tag. We also demonstrate metal-ion-induced protein phase separation in cells. The present phase separation strategy provides highly versatile protein condensates, which will greatly facilitate investigation of molecular and structural codes of droplet-forming proteins and the monitoring of biomolecular behaviors inside diverse protein condensates. Mimetic membrane-less organelles are of interest for the range of biochemical processes which can be spatio-temporally organized using them. Here, the authors report on a protein condensate system formed by metal ion induced clustering and demonstrate control over condensate properties.
Collapse
|
12
|
Bertran MT, Mouilleron S, Zhou Y, Bajaj R, Uliana F, Kumar GS, van Drogen A, Lee R, Banerjee JJ, Hauri S, O'Reilly N, Gstaiger M, Page R, Peti W, Tapon N. ASPP proteins discriminate between PP1 catalytic subunits through their SH3 domain and the PP1 C-tail. Nat Commun 2019; 10:771. [PMID: 30770806 PMCID: PMC6377682 DOI: 10.1038/s41467-019-08686-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 01/25/2019] [Indexed: 11/26/2022] Open
Abstract
Serine/threonine phosphatases such as PP1 lack substrate specificity and associate with a large array of targeting subunits to achieve the requisite selectivity. The tumour suppressor ASPP (apoptosis-stimulating protein of p53) proteins associate with PP1 catalytic subunits and are implicated in multiple functions from transcriptional regulation to cell junction remodelling. Here we show that Drosophila ASPP is part of a multiprotein PP1 complex and that PP1 association is necessary for several in vivo functions of Drosophila ASPP. We solve the crystal structure of the human ASPP2/PP1 complex and show that ASPP2 recruits PP1 using both its canonical RVxF motif, which binds the PP1 catalytic domain, and its SH3 domain, which engages the PP1 C-terminal tail. The ASPP2 SH3 domain can discriminate between PP1 isoforms using an acidic specificity pocket in the n-Src domain, providing an exquisite mechanism where multiple motifs are used combinatorially to tune binding affinity to PP1.
Collapse
Affiliation(s)
- M Teresa Bertran
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Stéphane Mouilleron
- Structural Biology - Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| | - Yanxiang Zhou
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Rakhi Bajaj
- Chemistry and Biochemistry Department, University of Arizona, 1041 E. Lowell Street, Biosciences West, 517, Tucson, AZ, 85721, USA
| | - Federico Uliana
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, 8093, Zürich, Switzerland
| | - Ganesan Senthil Kumar
- Chemistry and Biochemistry Department, University of Arizona, 1041 E. Lowell Street, Biosciences West, 517, Tucson, AZ, 85721, USA
| | - Audrey van Drogen
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, 8093, Zürich, Switzerland
| | - Rebecca Lee
- Structural Biology - Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Jennifer J Banerjee
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Simon Hauri
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, 8093, Zürich, Switzerland
| | - Nicola O'Reilly
- Peptide Chemistry Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, 8093, Zürich, Switzerland
| | - Rebecca Page
- Chemistry and Biochemistry Department, University of Arizona, 1041 E. Lowell Street, Biosciences West, 517, Tucson, AZ, 85721, USA
| | - Wolfgang Peti
- Chemistry and Biochemistry Department, University of Arizona, 1041 E. Lowell Street, Biosciences West, 517, Tucson, AZ, 85721, USA
| | - Nicolas Tapon
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
13
|
Vermeren S, Miles K, Chu JY, Salter D, Zamoyska R, Gray M. PTPN22 Is a Critical Regulator of Fcγ Receptor-Mediated Neutrophil Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:4771-4779. [PMID: 27807193 PMCID: PMC5136470 DOI: 10.4049/jimmunol.1600604] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 10/09/2016] [Indexed: 01/08/2023]
Abstract
Neutrophils act as a first line of defense against bacterial and fungal infections, but they are also important effectors of acute and chronic inflammation. Genome-wide association studies have established that the gene encoding the protein tyrosine phosphatase nonreceptor 22 (PTPN22) makes an important contribution to susceptibility to autoimmune disease, notably rheumatoid arthritis. Although PTPN22 is most highly expressed in neutrophils, its function in these cells remains poorly characterized. We show in this article that neutrophil effector functions, including adhesion, production of reactive oxygen species, and degranulation induced by immobilized immune complexes, were reduced in Ptpn22-/- neutrophils. Tyrosine phosphorylation of Lyn and Syk was altered in Ptpn22-/- neutrophils. On stimulation with immobilized immune complexes, Ptpn22-/- neutrophils manifested reduced activation of key signaling intermediates. Ptpn22-/- mice were protected from immune complex-mediated arthritis, induced by the transfer of arthritogenic serum. In contrast, in vivo neutrophil recruitment following thioglycollate-induced peritonitis and in vitro chemotaxis were not affected by lack of PTPN22. Our data suggest an important role for PTPN22-dependent dephosphorylation events, which are required to enable full FcγR-induced activation, pointing to an important role for this molecule in neutrophil function.
Collapse
Affiliation(s)
- Sonja Vermeren
- Medical Research Council/University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom;
| | - Katherine Miles
- Medical Research Council/University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Julia Y Chu
- Medical Research Council/University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Donald Salter
- Institute for Genetics and Molecular Medicine, Edinburgh EH4 2XU, United Kingdom; and
| | - Rose Zamoyska
- Institute of Immunology and Infection Research, Ashworth Laboratories, University of Edinburgh, Edinburgh EH9 3FL, United Kingdom
| | - Mohini Gray
- Medical Research Council/University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom;
| |
Collapse
|
14
|
Zhang R, Zhang J, Wu Q, Meng F, Liu C. XB130: A novel adaptor protein in cancer signal transduction. Biomed Rep 2016; 4:300-306. [PMID: 26998266 DOI: 10.3892/br.2016.588] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 01/18/2016] [Indexed: 12/13/2022] Open
Abstract
Adaptor proteins are functional proteins that contain two or more protein-binding modules to link signaling proteins together, which affect cell growth and shape and have no enzymatic activity. The actin filament-associated protein (AFAP) family is an important member of the adaptor proteins, including AFAP1, AFAP1L1 and AFAP1L2/XB130. AFAP1 and AFAP1L1 share certain common characteristics and function as an actin-binding protein and a cSrc-activating protein. XB130 exhibits certain unique features in structure and function. The mRNA of XB130 is expressed in human spleen, thyroid, kidney, brain, lung, pancreas, liver, colon and stomach, and the most prominent disease associated with XB130 is cancer. XB130 has a controversial effect on cancer. Studies have shown that XB130 can promote cancer progression and downregulation of XB130-reduced growth of tumors derived from certain cell lines. A higher mRNA level of XB130 was shown to be associated with a better survival in non-small cell lung cancer. Previous studies have shown that XB130 can regulate cell growth, migration and invasion and possibly has the effect through the cAMP-cSrc-phosphoinositide 3-kinase/Akt pathway. Except for cancer, XB130 is also associated with other pathological or physiological procedures, such as airway repair and regeneration.
Collapse
Affiliation(s)
- Ruiyao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Qifei Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Fandi Meng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| |
Collapse
|
15
|
|
16
|
Sarmiento J, Wallis RH, Ning T, Marandi L, Chao G, Veillette A, Lernmark Å, Paterson AD, Poussier P. A functional polymorphism of Ptpn22 is associated with type 1 diabetes in the BioBreeding rat. THE JOURNAL OF IMMUNOLOGY 2014; 194:615-29. [PMID: 25505293 DOI: 10.4049/jimmunol.1302689] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The R620W variant of PTPN22 is one of the major genetic risk factors for several autoimmune disorders including type 1 diabetes (T1D) in humans. In the BioBreeding T1D-prone (BBDP) rat, a single nucleotide polymorphism in Ptpn22 results in an A629T substitution immediately C-terminal to the aliphatic residues central to the Ptpn22-C-terminal Src kinase interaction. This variant exhibits a 50% decrease in C-terminal Src kinase binding affinity and contributes to T cell hyperresponsiveness. Examination of BBDP sublines congenic for the Iddm26.2 locus that includes Ptpn22 has not only shown an expansion of activated CD4(+)25(+) T lymphocytes in animals homozygous for the BBDP allele, consistent with enhanced TCR-mediated signaling, but also a decrease in their proportion of peripheral Foxp3(+) regulatory T cells. Furthermore, clinical assessment of both an F2(BBDP × ACI.1u.Lyp) cohort and Iddm26.2 congenic BBDP sublines has revealed an association of Ptpn22 with T1D. Specifically, in both cases, T1D risk is significantly greater in BBDP Ptpn22 homozygous and heterozygous animals. These findings are consistent with a role for rat Ptpn22 allelic variation within Iddm26.2 in the regulation of T cell responses, and subsequently the risk for development of T1D.
Collapse
Affiliation(s)
- Janice Sarmiento
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Robert H Wallis
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Terri Ning
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Leili Marandi
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Gary Chao
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - André Veillette
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec H2W 1R7, Canada
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden
| | - Andrew D Paterson
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario M5T 3M7, Canada; and Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Philippe Poussier
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada;
| |
Collapse
|
17
|
He RJ, Yu ZH, Zhang RY, Zhang ZY. Protein tyrosine phosphatases as potential therapeutic targets. Acta Pharmacol Sin 2014; 35:1227-46. [PMID: 25220640 DOI: 10.1038/aps.2014.80] [Citation(s) in RCA: 262] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/31/2014] [Indexed: 12/17/2022]
Abstract
Protein tyrosine phosphorylation is a key regulatory process in virtually all aspects of cellular functions. Dysregulation of protein tyrosine phosphorylation is a major cause of human diseases, such as cancers, diabetes, autoimmune disorders, and neurological diseases. Indeed, protein tyrosine phosphorylation-mediated signaling events offer ample therapeutic targets, and drug discovery efforts to date have brought over two dozen kinase inhibitors to the clinic. Accordingly, protein tyrosine phosphatases (PTPs) are considered next-generation drug targets. For instance, PTP1B is a well-known targets of type 2 diabetes and obesity, and recent studies indicate that it is also a promising target for breast cancer. SHP2 is a bona-fide oncoprotein, mutations of which cause juvenile myelomonocytic leukemia, acute myeloid leukemia, and solid tumors. In addition, LYP is strongly associated with type 1 diabetes and many other autoimmune diseases. This review summarizes recent findings on several highly recognized PTP family drug targets, including PTP1B, Src homology phosphotyrosyl phosphatase 2(SHP2), lymphoid-specific tyrosine phosphatase (LYP), CD45, Fas associated phosphatase-1 (FAP-1), striatal enriched tyrosine phosphatases (STEP), mitogen-activated protein kinase/dual-specificity phosphatase 1 (MKP-1), phosphatases of regenerating liver-1 (PRL), low molecular weight PTPs (LMWPTP), and CDC25. Given that there are over 100 family members, we hope this review will serve as a road map for innovative drug discovery targeting PTPs.
Collapse
|
18
|
Effects of protein tyrosine phosphatase-PEST are reversed by Akt in T cells. Cell Signal 2014; 26:2721-9. [PMID: 25152368 DOI: 10.1016/j.cellsig.2014.08.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/17/2014] [Indexed: 11/21/2022]
Abstract
T cell activation is regulated by a balance between phosphorylation and dephosphorylation that is under the control of kinases and phosphatases. Here, we examined the role of a non-receptor-type protein tyrosine phosphatase, PTP-PEST, using retrovirus-mediated gene transduction into murine T cells. Based on observations of vector markers (GFP or Thy1.1), exogenous PTP-PEST-positive CD4(+) T cells appeared within 2 days after gene transduction; the percentage of PTP-PEST-positive cells tended to decrease during a resting period in the presence of IL-2 over the next 2 days. These vector markers also showed much lower expression intensities, compared with control cells, suggesting a correlation between the percent reduction and the low marker expression intensity. A catalytically inactive PTP-PEST mutant also showed the same tendency, and stepwise deletion mutants gradually lost their ability to induce the above phenomenon. On the other hand, these PTP-PEST-transduced cells did not have an apoptotic phenotype. No difference in the total cell numbers was found in the wells of a culture plate containing VEC- and PTP-PEST-transduced T cells. Moreover, serine/threonine kinase Akt, but not the anti-apoptotic molecules Bcl-2 and Bcl-XL, reversed the phenotype induced by PTP-PEST. We discuss the novel mechanism by which Akt interferes with PTP-PEST.
Collapse
|
19
|
Gianchecchi E, Palombi M, Fierabracci A. The putative role of the C1858T polymorphism of protein tyrosine phosphatase PTPN22 gene in autoimmunity. Autoimmun Rev 2013; 12:717-725. [PMID: 23261816 DOI: 10.1016/j.autrev.2012.12.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 12/05/2012] [Indexed: 12/11/2022]
Abstract
Autoimmune diseases represent a heterogeneous group of conditions whose incidence is increasing worldwide. This has stimulated studies on their etiopathogenesis, derived from a complex interaction between genetic and environmental factors, in order to improve prevention and treatment of these diseases. An increasing amount of epidemiologic investigations has associated the presence of the C1858T polymorphism in the protein tyrosine phosphatase non-receptor type 22 (PTPN22) gene to the onset of several autoimmune diseases including insulin-dependent diabetes mellitus (Type 1 diabetes). PTPN22 encodes for the lymphoid tyrosine phosphatase Lyp. This belongs to non-receptor-type protein tyrosine phosphatases involved in lymphocyte activation and differentiation. In humans, Lyp may have a role in the negative regulation of T cell receptor signaling. The single nucleotide polymorphism C1858T encodes for a more active phosphatase Lyp R620W. This has the ability to induce a higher negative regulation of T cell receptor signaling. Thus, C1858T could play an important role at the level of thymocyte polarization and escape of autoreactive T lymphocytes, through the positive selection of otherwise negatively selected autoimmune T cells. In this review we discuss the physiological role exerted by the PTPN22 gene and its encoded Lyp product in lymphocyte processes. We highlight the pathogenic significance of the C1858T PTPN22 polymorphism in human autoimmunity with special reference to Type 1 diabetes. Recently the genetic variation in PTPN22 was shown to induce altered function of T and B-lymphocytes. In particular BCR signaling defects and alterations in the B cell compartment were reported in T1D patients. We finally speculate on the possible development of novel therapeutic treatments in human autoimmunity aiming to selectively target the variant Lyp protein in autoreactive T and B lymphocytes.
Collapse
Affiliation(s)
- Elena Gianchecchi
- Autoimmunity Laboratory, Immunology Area, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | | | | |
Collapse
|
20
|
de la Puerta ML, Trinidad AG, Rodríguez MDC, de Pereda JM, Sánchez Crespo M, Bayón Y, Alonso A. The autoimmunity risk variant LYP-W620 cooperates with CSK in the regulation of TCR signaling. PLoS One 2013; 8:e54569. [PMID: 23359562 PMCID: PMC3554717 DOI: 10.1371/journal.pone.0054569] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 12/12/2012] [Indexed: 11/21/2022] Open
Abstract
The protein tyrosine phosphatase LYP, a key regulator of TCR signaling, presents a single nucleotide polymorphism, C1858T, associated with several autoimmune diseases such as type I diabetes, rheumatoid arthritis, and lupus. This polymorphism changes an R by a W in the P1 Pro rich motif of LYP, which binds to CSK SH3 domain, another negative regulator of TCR signaling. Based on the analysis of the mouse homologue, Pep, it was proposed that LYP and CSK bind constitutively to inhibit LCK and subsequently TCR signaling. The detailed study of LYP/CSK interaction, here presented, showed that LYP/CSK interaction was inducible upon TCR stimulation, and involved LYP P1 and P2 motifs, and CSK SH3 and SH2 domains. Abrogating LYP/CSK interaction did not preclude the regulation of TCR signaling by these proteins.
Collapse
Affiliation(s)
- María Luisa de la Puerta
- Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Antonio G. Trinidad
- Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, Valladolid, Spain
| | | | - José María de Pereda
- Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
| | - Mariano Sánchez Crespo
- Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Yolanda Bayón
- Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Andrés Alonso
- Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, Valladolid, Spain
- * E-mail:
| |
Collapse
|
21
|
Stanford SM, Rapini N, Bottini N. Regulation of TCR signalling by tyrosine phosphatases: from immune homeostasis to autoimmunity. Immunology 2012; 137:1-19. [PMID: 22862552 DOI: 10.1111/j.1365-2567.2012.03591.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
More than half of the known protein tyrosine phosphatases (PTPs) in the human genome are expressed in T cells, and significant progress has been made in elucidating the biology of these enzymes in T-cell development and function. Here we provide a systematic review of the current understanding of the roles of PTPs in T-cell activation, providing insight into their mechanisms of action and regulation in T-cell receptor signalling, the phenotypes of their genetically modified mice, and their possible involvement in T-cell-mediated autoimmune disease. Our projection is that the interest in PTPs as mediators of T-cell homeostasis will continue to rise with further functional analysis of these proteins, and PTPs will be increasingly considered as targets of immunomodulatory therapies.
Collapse
Affiliation(s)
- Stephanie M Stanford
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
22
|
SH3 domains: modules of protein-protein interactions. Biophys Rev 2012; 5:29-39. [PMID: 28510178 DOI: 10.1007/s12551-012-0081-z] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 05/29/2012] [Indexed: 01/01/2023] Open
Abstract
Src homology 3 (SH3) domains are involved in the regulation of important cellular pathways, such as cell proliferation, migration and cytoskeletal modifications. Recognition of polyproline and a number of noncanonical sequences by SH3 domains has been extensively studied by crystallography, nuclear magnetic resonance and other methods. High-affinity peptides that bind SH3 domains are used in drug development as candidates for anticancer treatment. This review summarizes the latest achievements in deciphering structural determinants of SH3 function.
Collapse
|
23
|
Rhee I, Veillette A. Protein tyrosine phosphatases in lymphocyte activation and autoimmunity. Nat Immunol 2012; 13:439-47. [PMID: 22513334 DOI: 10.1038/ni.2246] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lymphocyte activation must be tightly regulated to ensure sufficient immunity to pathogens and prevent autoimmunity. Protein tyrosine phosphatases (PTPs) serve critical roles in this regulation by controlling the functions of key receptors and intracellular signaling molecules in lymphocytes. In some cases, PTPs inhibit lymphocyte activation, whereas in others they promote it. Here we discuss recent progress in elucidating the roles and mechanisms of action of PTPs in lymphocyte activation. We also review the accumulating evidence that genetic alterations in PTPs are involved in human autoimmunity.
Collapse
Affiliation(s)
- Inmoo Rhee
- Laboratory of Molecular Oncology, Clinical Research Institute of Montréal, Montréal, Québec, Canada
| | | |
Collapse
|
24
|
Vang T, Liu WH, Delacroix L, Wu S, Vasile S, Dahl R, Yang L, Musumeci L, Francis D, Landskron J, Tasken K, Tremblay ML, Lie BA, Page R, Mustelin T, Rahmouni S, Rickert RC, Tautz L. LYP inhibits T-cell activation when dissociated from CSK. Nat Chem Biol 2012; 8:437-46. [PMID: 22426112 PMCID: PMC3329573 DOI: 10.1038/nchembio.916] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 01/27/2012] [Indexed: 01/17/2023]
Abstract
Lymphoid tyrosine phosphatase (LYP) and C-terminal Src kinase (CSK) are negative regulators of signaling mediated through the T cell antigen receptor (TCR) and are thought to act in a cooperative manner when forming a complex. Here, we studied the spatio-temporal dynamics of the LYP/CSK complex in T cells. We demonstrate that dissociation of this complex is necessary for recruitment of LYP to the plasma membrane, where it down-modulates TCR signaling. Development of a potent and selective chemical probe of LYP confirmed that LYP inhibits T cell activation when removed from CSK. Our findings may explain the reduced TCR-mediated signaling associated with a single nucleotide polymorphism, which confers increased risk for certain autoimmune diseases, including type 1 diabetes and rheumatoid arthritis, and results in expression of a LYP allele that is unable to bind CSK. Our compound also represents a starting point for the development of a LYP-based treatment of autoimmunity.
Collapse
Affiliation(s)
- Torkel Vang
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Polychronakos C, Li Q. Understanding type 1 diabetes through genetics: advances and prospects. Nat Rev Genet 2011; 12:781-92. [PMID: 22005987 DOI: 10.1038/nrg3069] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Starting with early crucial discoveries of the role of the major histocompatibility complex, genetic studies have long had a role in understanding the biology of type 1 diabetes (T1D), which is one of the most heritable common diseases. Recent genome-wide association studies (GWASs) have given us a clearer picture of the allelic architecture of genetic susceptibility to T1D. Fine mapping and functional studies are gradually revealing the complex mechanisms whereby immune self-tolerance is lost, involving multiple aspects of adaptive immunity. The triggering of these events by dysregulation of the innate immune system has also been implicated by genetic evidence. Finally, genetic prediction of T1D risk is showing promise of use for preventive strategies.
Collapse
Affiliation(s)
- Constantin Polychronakos
- Departments of Pediatrics and Human Genetics, McGill University, Montreal, Québec, Canada H3H 1P3. Constantin.
| | | |
Collapse
|
26
|
The autoimmune disease-associated PTPN22 variant promotes calpain-mediated Lyp/Pep degradation associated with lymphocyte and dendritic cell hyperresponsiveness. Nat Genet 2011; 43:902-7. [PMID: 21841778 DOI: 10.1038/ng.904] [Citation(s) in RCA: 206] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 07/14/2011] [Indexed: 12/27/2022]
Abstract
A variant of the PTPN22-encoded Lyp phosphatase (Lyp620W) confers risk for autoimmune disease, but the mechanisms underlying this association remain unclear. We show here that mice expressing the Lyp variant homolog Pep619W manifest thymic and splenic enlargement accompanied by increases in T-cell number, activation and positive selection and in dendritic- and B-cell activation. Although Ptpn22 (Pep) transcript levels were comparable in Pep619W and wild-type Pep619R mice, Pep protein levels were dramatically reduced in the mutant mice, with Pep619W protein being more rapidly degraded and showing greater association with and in vitro cleavage by calpain 1 than Pep619R. Similarly, levels of the Lyp620W variant were decreased in human T and B cells, and its calpain binding and cleavage were increased relative to wild-type Lyp620R. Thus, calpain-mediated degradation with consequently reduced Lyp/Pep expression and lymphocyte and dendritic cell hyperresponsiveness represents a mechanism whereby Lyp620W may increase risk for autoimmune disease.
Collapse
|
27
|
Burn GL, Svensson L, Sanchez-Blanco C, Saini M, Cope AP. Why is PTPN22 a good candidate susceptibility gene for autoimmune disease? FEBS Lett 2011; 585:3689-98. [PMID: 21515266 DOI: 10.1016/j.febslet.2011.04.032] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 04/13/2011] [Accepted: 04/13/2011] [Indexed: 10/18/2022]
Abstract
The PTPN22 locus is one of the strongest risk factors outside of the major histocompatability complex that associates with autoimmune diseases. PTPN22 encodes lymphoid protein tyrosine phosphatase (Lyp) which is expressed exclusively in immune cells. A single base change in the coding region of this gene resulting in an arginine to tryptophan amino acid substitution within a polyproline binding motif associates with type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosis, Hashimotos thyroiditis, Graves disease, Addison's disease, Myasthenia Gravis, vitiligo, systemic sclerosis juvenile idiopathic arthritis and psoriatic arthritis. Here, we review the current understanding of the PTPN22 locus from a genetic, geographical, biochemical and functional perspective.
Collapse
Affiliation(s)
- Garth L Burn
- Academic Department of Rheumatology, Division of Immunology, Infection and Inflammatory Disease, King's College School of Medicine, King's College London, UK.
| | | | | | | | | |
Collapse
|
28
|
Wang S, Dong H, Han J, Ho WT, Fu X, Zhao ZJ. Identification of a variant form of tyrosine phosphatase LYP. BMC Mol Biol 2010; 11:78. [PMID: 21044313 PMCID: PMC2987843 DOI: 10.1186/1471-2199-11-78] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 11/02/2010] [Indexed: 01/27/2023] Open
Abstract
Background Protein tyrosine phosphatases (PTPs) are important cell signaling regulators with major pathological implications. LYP (also known as PTPN22) is an intracellular enzyme initially found to be predominately expressed in lymphocytes. Importantly, an allelic R620W variant of LYP is strongly associated with multiple autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes, and autoimmune thyroid disease. Results In this study, we isolated a novel isoform of LYP designated LYP3. LYP3 differs from LYP1, the known isoform of LYP, in that it lacks a 28 amino acid segment right after the R620W site embedded in a proline-rich protein-protein interaction motif. Genomic sequence analysis revealed that LYP3 resulted from alternative splicing of the LYP gene located on chromosome 1p 13.3-13.1. Reverse transcription PCR analyses of 48 human tissues demonstrated that both LYP1 and LYP3 are predominantly expressed in primary and secondary lymphoid tissues but the relative expression levels of the two isoforms varies in different human tissues and individuals. Conclusions We thus identified a new variant form of LYP and conducted a comprehensive analysis of LYP tissue expressions. Considering the pathogenesis of LYP R620W, we believe that the expression of LYP3 may have an important role in regulating activity and function of LYP and may be implicated in autoimmune diseases.
Collapse
Affiliation(s)
- Shaofeng Wang
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | | | |
Collapse
|
29
|
Stanford SM, Mustelin TM, Bottini N. Lymphoid tyrosine phosphatase and autoimmunity: human genetics rediscovers tyrosine phosphatases. Semin Immunopathol 2010; 32:127-36. [PMID: 20204370 PMCID: PMC2881161 DOI: 10.1007/s00281-010-0201-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Accepted: 01/28/2010] [Indexed: 01/22/2023]
Abstract
A relatively large number of protein tyrosine phosphatases (PTPs) are known to regulate signaling through the T cell receptor (TCR). Recent human genetics studies have shown that several of these PTPs are encoded by major autoimmunity genes. Here, we will focus on the lymphoid tyrosine phosphatase (LYP), a critical negative modulator of TCR signaling encoded by the PTPN22 gene. The functional analysis of autoimmune-associated PTPN22 genetic variants suggests that genetic variability of TCR signal transduction contributes to the pathogenesis of autoimmunity in humans.
Collapse
Affiliation(s)
- Stephanie M. Stanford
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Tomas M. Mustelin
- Sanford-Burnham Medical Research Institute, Infectious and Inflammatory Disease Center, La Jolla, CA 92037, USA
| | - Nunzio Bottini
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA.
| |
Collapse
|
30
|
Samet JM, Tal TL. Toxicological disruption of signaling homeostasis: tyrosine phosphatases as targets. Annu Rev Pharmacol Toxicol 2010; 50:215-35. [PMID: 20055703 DOI: 10.1146/annurev.pharmtox.010909.105841] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The protein tyrosine phosphatases (PTPs) consist of a diverse group of enzymes whose activity opposes that of the tyrosine kinases. As such, the PTPs have critical roles in maintaining signaling quiescence in resting cells and in restoring homeostasis by effecting signal termination. Interest in these enzymes has increased in recent years following the discovery that the activity of PTPs is modulated through redox mechanisms during signaling. The molecular features that enable redox regulation of PTPs during physiological signaling also render them highly susceptible to oxidative and electrophilic inactivation by a broad spectrum of structurally disparate xenobiotic compounds. The loss of PTP activity results in a profound disregulation of protein phosphotyrosine metabolism, leading to widespread and persistent activation of signaling cascades in the cell.
Collapse
Affiliation(s)
- James M Samet
- U.S. Environmental Protection Agency, Chapel Hill, North Carolina, USA.
| | | |
Collapse
|
31
|
Abstract
The Src family kinases possess two sites of tyrosine phosphorylation that are critical to the regulation of kinase activity. Autophosphorylation on an activation loop tyrosine residue (Tyr 416 in commonly used chicken c-Src numbering) increases catalytic activity, while phosphorylation of a C-terminal tyrosine (Tyr 527 in c-Src) inhibits activity. The latter modification is achieved by the tyrosine kinase Csk (C-terminal Src Kinase), but the complete inactivation of the Src family kinases also requires the dephosphorylation of the activation loop tyrosine. The SH3 domain of Csk recruits the tyrosine phosphatase PEP, allowing for the coordinated inhibition of Src family kinase activity. We have discovered that Csk forms homodimers through interactions mediated by the SH3 domain in a manner that buries the recognition surface for SH3 ligands. The formation of this dimer would therefore block the recruitment of tyrosine phosphatases and may have important implications for the regulation of Src kinase activity.
Collapse
|
32
|
Li Y, Begovich AB. Unraveling the genetics of complex diseases: susceptibility genes for rheumatoid arthritis and psoriasis. Semin Immunol 2009; 21:318-27. [PMID: 19446472 DOI: 10.1016/j.smim.2009.04.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 04/09/2009] [Indexed: 12/29/2022]
Abstract
Talk of numerous genetic risk factors for rheumatoid arthritis (RA) and psoriasis has been percolating for years, but with the exception of the human leukocyte antigen (HLA) region, none have been definitively identified. Recently the results of multiple, well powered, genetic case-control studies have begun to appear providing convincing statistical evidence for at least ten non-HLA related risk genes or loci (C5/TRAF1, CD40, CTLA4, KIF5A/PIP4K2C, MMEL1/TNFRSF14, PADI4, PRKCQ, PTPN22, STAT4, and TNFAIP3/OLIG3) for RA and six (IL12B, IL13, IL23R, STAT2/IL23A, TNFAIP3, and TNIP1) for psoriasis. These initial, novel findings are beginning to shed light on the molecular pathways pertinent to the individual diseases and highlight the pleiotropic effects of several risk factors as well as the allelic heterogeneity underlying susceptibility to these and other autoimmune diseases.
Collapse
Affiliation(s)
- Yonghong Li
- Celera, 1401 Harbor Bay Parkway, Alameda, CA 94502, USA
| | | |
Collapse
|
33
|
Veillette A, Rhee I, Souza CM, Davidson D. PEST family phosphatases in immunity, autoimmunity, and autoinflammatory disorders. Immunol Rev 2009; 228:312-24. [PMID: 19290936 DOI: 10.1111/j.1600-065x.2008.00747.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The proline-, glutamic acid-, serine- and threonine-rich (PEST) family of protein tyrosine phosphatases (PTPs) includes proline-enriched phosphatase (PEP)/lymphoid tyrosine phosphatase (LYP), PTP-PEST, and PTP-hematopoietic stem cell fraction (HSCF). PEP/LYP is a potent inhibitor of T-cell activation, principally by suppressing the activity of Src family protein tyrosine kinases (PTKs). This function seems to be dependent, at least in part, on the ability of PEP to bind C-terminal Src kinase (Csk), a PTK also involved in inactivating Src kinases. Interestingly, a polymorphism of LYP in humans (R620W) is a significant risk factor for autoimmune diseases including type 1 diabetes, rheumatoid arthritis, and lupus. The R620W mutation may be a 'gain-of-function' mutation. In non-hematopoietic cells, PTP-PEST is a critical regulator of adhesion and migration. This effect correlates with the aptitude of PTP-PEST to dephosphorylate cytoskeletal proteins such as Cas, focal adhesion associated-kinase (FAK), Pyk2, and PSTPIP. While not established, a similar function may also exist in immune cells. Additionally, overexpression studies provided an indication that PTP-PEST may be a negative regulator of lymphocyte activation. Interestingly, mutations in a PTP-PEST- and PTP-HSCF-interacting protein, PSTPIP1, were identified in humans with pyogenic sterile arthritis, pyoderma gangrenosum, and acne (PAPA) syndrome and familial recurrent arthritis, two autoinflammatory diseases. These mutations abrogate the ability of PSTPIP1 to bind PTP-PEST and PTP-HSCF, suggesting that these two PTPs may be negative regulators of inflammation.
Collapse
Affiliation(s)
- André Veillette
- Laboratory of Molecular Oncology, Clinical Research Institute of Montréal, Montréal, Québec, Canada.
| | | | | | | |
Collapse
|
34
|
Hermiston ML, Zikherman J, Zhu JW. CD45, CD148, and Lyp/Pep: critical phosphatases regulating Src family kinase signaling networks in immune cells. Immunol Rev 2009; 228:288-311. [PMID: 19290935 PMCID: PMC2739744 DOI: 10.1111/j.1600-065x.2008.00752.x] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Reciprocal regulation of tyrosine phosphorylation by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) is central to normal immune cell function. Disruption of the equilibrium between PTK and PTP activity can result in immunodeficiency, autoimmunity, or malignancy. Src family kinases (SFKs) play a central role in both immune cell function and disease due to their proximal position in numerous signal transduction cascades including those emanating from integrin, T and B-cell antigen receptors, Fc, growth factor, and cytokine receptors. Given that tight regulation of SFKs activity is critical for appropriate responses to stimulation of these various signaling pathways, it is perhaps not surprising that multiple PTPs are involved in their regulation. Here, we focus on the role of three phosphatases, CD45, CD148, and LYP/PEP, which are critical regulators of SFKs in hematopoietic cells. We review our current understanding of their structures, expression, functions in different hematopoietic cell subsets, regulation, and putative roles in disease. Finally, we discuss remaining questions that must be addressed if we are to have a clearer understanding of the coordinated regulation of tyrosine phosphorylation and signaling networks in hematopoietic cells and how they could potentially be manipulated therapeutically in disease.
Collapse
Affiliation(s)
- Michelle L. Hermiston
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, Phone: 415-476-2413, Fax: 415-502-5127,
| | - Julie Zikherman
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, Phone: 415-476-4115, Fax: 502-5081, ;
| | - Jing W. Zhu
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, Phone: 415-476-4115, Fax: 502-5081, ;
| |
Collapse
|
35
|
Phelan JD, Thompson SD, Glass DN. Susceptibility to JRA/JIA: complementing general autoimmune and arthritis traits. Genes Immun 2009; 7:1-10. [PMID: 16435022 DOI: 10.1038/sj.gene.6364273] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Juvenile rheumatoid arthritis (JRA), also known as juvenile idiopathic arthritis (JIA), includes the most common chronic autoimmune arthropathies of childhood. These two nomenclatures for classification include components representing the major subclasses of disease. The chromosomal regions and the genes involved in these complex genetic traits are being elucidated, with findings often specific for a particular disease subtype. With the advent of new SNP technologies, progress is being made at an ever-increasing pace. This review discusses the difficulties of deciphering the genetic components in complex disorders, while demonstrating the similarities that JRA shares with other autoimmune disorders. Particular emphasis has been placed on positive findings either for candidate genes that have been replicated independently in JRA/JIA, or findings in JRA for which consistent results have been reported in other forms of autoimmunity.
Collapse
Affiliation(s)
- J D Phelan
- William S Rowe Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | | | | |
Collapse
|
36
|
Vang T, Miletic AV, Arimura Y, Tautz L, Rickert RC, Mustelin T. Protein tyrosine phosphatases in autoimmunity. Annu Rev Immunol 2008; 26:29-55. [PMID: 18303998 DOI: 10.1146/annurev.immunol.26.021607.090418] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein tyrosine phosphatases (PTPs) are important regulators of many cellular functions and a growing number of PTPs have been implicated in human disease conditions, such as developmental defects, neoplastic disorders, and immunodeficiency. Here, we review the involvement of PTPs in human autoimmunity. The leading examples include the allelic variant of the lymphoid tyrosine phosphatase (PTPN22), which is associated with multiple autoimmune diseases, and mutations that affect the exon-intron splicing of CD45 (PTPRC). We also find it likely that additional PTPs are involved in susceptibility to autoimmune and inflammatory diseases. Finally, we discuss the possibility that PTPs regulating the immune system may serve as therapeutic targets.
Collapse
Affiliation(s)
- Torkel Vang
- Burnham Institute for Medical Research, La Jolla, California 92037, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Kadaveru K, Vyas J, Schiller MR. Viral infection and human disease--insights from minimotifs. FRONTIERS IN BIOSCIENCE : A JOURNAL AND VIRTUAL LIBRARY 2008; 13:6455-71. [PMID: 18508672 PMCID: PMC2628544 DOI: 10.2741/3166] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Short functional peptide motifs cooperate in many molecular functions including protein interactions, protein trafficking, and posttranslational modifications. Viruses exploit these motifs as a principal mechanism for hijacking cells and many motifs are necessary for the viral life-cycle. A virus can accommodate many short motifs in its small genome size providing a plethora of ways for the virus to acquire host molecular machinery. Host enzymes that act on motifs such as kinases, proteases, and lipidation enzymes, as well as protein interaction domains, are commonly mutated in human disease, suggesting that the short peptide motif targets of these enzymes may also be mutated in disease; however, this is not observed. How can we explain why viruses have evolved to be so dependent on motifs, yet these motifs, in general do not seem to be as necessary for human viability? We propose that short motifs are used at the system level. This system architecture allows viruses to exploit a motif, whereas the viability of the host is not affected by mutation of a single motif.
Collapse
Affiliation(s)
- Krishna Kadaveru
- University of Connecticut Health Center, Department of Molecular, Microbial, and Structural Biology, Biological Systems Modeling Group, 263 Farmington Ave., Farmington, CT, 06030-3305
| | - Jay Vyas
- University of Connecticut Health Center, Department of Molecular, Microbial, and Structural Biology, Biological Systems Modeling Group, 263 Farmington Ave., Farmington, CT, 06030-3305
| | - Martin R. Schiller
- University of Connecticut Health Center, Department of Molecular, Microbial, and Structural Biology, Biological Systems Modeling Group, 263 Farmington Ave., Farmington, CT, 06030-3305
| |
Collapse
|
38
|
Abstract
The discovery that a single-nucleotide polymorphism (SNP) in lymphoid tyrosine phosphatase (LYP), encoded by the PTPN22 gene, is associated with type 1 diabetes (T1D) has now been verified by numerous studies and has been expanded to rheumatoid arthritis, juvenile rheumatoid arthritis (JRA), systemic lupus erythematosus, Graves' disease, generalized vitiligo and other human autoimmune diseases. In this paper, we discuss the association of PTPN22 with autoimmunity, the biochemistry of the PTPN22-encoded phosphatase, and the molecular mechanism(s) by which the disease-predisposing allele contributes to the development of human disease.
Collapse
MESH Headings
- Alleles
- Autoimmune Diseases/enzymology
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Autoimmunity
- Genetic Predisposition to Disease
- Humans
- Polymorphism, Single Nucleotide
- Protein Tyrosine Phosphatase, Non-Receptor Type 22/chemistry
- Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Torkel Vang
- The Burnham Institute for Medical Research, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
39
|
Xu J, Bai XH, Lodyga M, Han B, Xiao H, Keshavjee S, Hu J, Zhang H, Yang BB, Liu M. XB130, a Novel Adaptor Protein for Signal Transduction. J Biol Chem 2007; 282:16401-12. [PMID: 17412687 DOI: 10.1074/jbc.m701684200] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Adaptor proteins are important mediators in signal transduction. In the present study, we report the cloning and characterization of a novel adaptor protein, XB130. This gene is located on human chromosome 10q25.3 and encodes a protein of 818 amino acids. It contains several Src homology (SH)2- and SH3-binding motifs, two pleckstrin homology domains, a coiled-coil region, and a number of potential tyrosine or serine/threonine phosphorylation sites. Endogenous XB130 interacts with c-Src tyrosine kinase. Their co-expression in COS-7 cells resulted in activation of c-Src and elevated tyrosine phosphorylation of multiple proteins, including XB130 itself. XB130 expression in HEK293 cells enhanced serum response element- and AP-1-dependent transcriptional activation mediated by c-Src. XB130DeltaN, an N-terminal deletion mutant lacking a putative SH3-binding motif and several putative SH2-binding sites, reduced its ability to mediate Src signal transduction. Down-regulation of endogenous XB130 with siRNA reduced c-Src activity, IL-8 production, EGF-induced phosphorylation of Akt and GSK3beta, and altered cell cycles in human lung epithelial cells. These data suggest that XB130 as an adaptor may play an important role in the regulation of signal transduction and cellular functions.
Collapse
Affiliation(s)
- Jing Xu
- Division of Cellular and Molecular Biology, University Health Network Toronto General Research Institute, and Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Tyrosyl phosphorylation plays a critical role in multiple signaling pathways regulating innate and acquired immunity. Although tyrosyl phosphorylation is a reversible process, we know much more about the functions of protein-tyrosine kinases (PTKs) than about protein-tyrosine phosphatases (PTPs). Genome sequencing efforts have revealed a large and diverse superfamily of PTPs, which can be subdivided into receptor-like (RPTPs) and nonreceptor (NRPTPs). The role of the RPTP CD45 in immune cell signaling is well known, but those of most other PTPs remain poorly understood. Here, we review the mechanism of action, regulation, and physiological functions of NRPTPs in immune cell signaling. Such an analysis indicates that PTPs are as important as PTKs in regulating the immune system.
Collapse
Affiliation(s)
- Lily I Pao
- Cancer Biology Program, Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA.
| | | | | | | |
Collapse
|
41
|
Abstract
We recently discovered that a single-nucleotide polymorphism (SNP) in the lymphoid tyrosine phosphatase (LYP), encoded by the PTPN22 gene on chromosome 1p13, correlates strongly with the incidence of type 1 diabetes (T1D) in two independent populations. This findings has now been verified by numerous studies and it has been expanded to rheumatoid arthritis, juvenile rheumatoid arthritis, systemic lupus erythematosus, Graves' disease, generalized vitiligo and other autoimmune disease. Here we review the genetics of the SNP and its association with autoimmunity, discuss the function of the phosphatase in signaling, the biochemistry of the disease-predisposing allele, and the possible mechanisms by which PTPN22 contributes to the development of human disease.
Collapse
Affiliation(s)
- Nunzio Bottini
- Institute for Genetic Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, United States
| | | | | | | |
Collapse
|
42
|
Carlton VEH, Hu X, Chokkalingam AP, Schrodi SJ, Brandon R, Alexander HC, Chang M, Catanese JJ, Leong DU, Ardlie KG, Kastner DL, Seldin MF, Criswell LA, Gregersen PK, Beasley E, Thomson G, Amos CI, Begovich AB. PTPN22 genetic variation: evidence for multiple variants associated with rheumatoid arthritis. Am J Hum Genet 2005; 77:567-81. [PMID: 16175503 PMCID: PMC1275606 DOI: 10.1086/468189] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2005] [Accepted: 07/19/2005] [Indexed: 01/29/2023] Open
Abstract
The minor allele of the R620W missense single-nucleotide polymorphism (SNP) (rs2476601) in the hematopoietic-specific protein tyrosine phosphatase gene, PTPN22, has been associated with multiple autoimmune diseases, including rheumatoid arthritis (RA). These genetic data, combined with biochemical evidence that this SNP affects PTPN22 function, suggest that this phosphatase is a key regulator of autoimmunity. To determine whether other genetic variants in PTPN22 contribute to the development of RA, we sequenced the coding regions of this gene in 48 white North American patients with RA and identified 15 previously unreported SNPs, including 2 coding SNPs in the catalytic domain. We then genotyped 37 SNPs in or near PTPN22 in 475 patients with RA and 475 individually matched controls (sample set 1) and selected a subset of markers for replication in an additional 661 patients with RA and 1,322 individually matched controls (sample set 2). Analyses of these results predict 10 common (frequency >1%) PTPN22 haplotypes in white North Americans. The sole haplotype found to carry the previously identified W620 risk allele was strongly associated with disease in both sample sets, whereas another haplotype, identical at all other SNPs but carrying the R620 allele, showed no association. R620W, however, does not fully explain the association between PTPN22 and RA, since significant differences between cases and controls persisted in both sample sets after the haplotype data were stratified by R620W. Additional analyses identified two SNPs on a single common haplotype that are associated with RA independent of R620W, suggesting that R620W and at least one additional variant in the PTPN22 gene region influence RA susceptibility.
Collapse
Affiliation(s)
- Victoria E. H. Carlton
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Xiaolan Hu
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Anand P. Chokkalingam
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Steven J. Schrodi
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Rhonda Brandon
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Heather C. Alexander
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Monica Chang
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Joseph J. Catanese
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Diane U. Leong
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Kristin G. Ardlie
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Daniel L. Kastner
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Michael F. Seldin
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Lindsey A. Criswell
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Peter K. Gregersen
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Ellen Beasley
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Glenys Thomson
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Christopher I. Amos
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| | - Ann B. Begovich
- Celera Diagnostics, Alameda, CA; Celera Genomics, Rockville, MD; Genomics Collaborative Division of SeraCare Life Sciences, Cambridge, MA; Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD; Rowe Program of Human Genetics, Department of Medicine, University of California–Davis, Davis; Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California–San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore–Long Island Jewish Institute for Medical Research, Manhasset, NY; Department of Integrative Biology, University of California–Berkeley, Berkeley; and Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, University of Texas, Houston
| |
Collapse
|
43
|
Abstract
Rheumatoid arthritis (RA), like other autoimmune diseases, has a complex genetic basis. Rapid technical advances in high-throughput genotyping and analysis have now reached a point where genes of low-to-moderate risk can be identified using a variety of study designs, including whole genome association studies. The availability of large, well-characterized populations of cases and controls are critical to the success of these efforts. A functional variant (R620W) of the intracellular protein tyrosine phosphatase N22 (PTPN22) has now been conclusively shown to confer approximately two-fold risk for seropositive RA as well as several other autoimmune disorders. PTPN22 appears to act primarily by setting thresholds for T-cell receptor signaling, and the current data suggest that the PTPN22 620W allele is likely to be a general risk factor for the development of humoral autoimmunity. PTPN22 is expressed widely in hematopoietic cells, but other than in T cells, its role is unknown. These results provide strong evidence for the longstanding hypothesis that common genes underlie different autoimmune phenotypes and emphasize that finding genes of only moderate risk can provide important insights into disease pathogenesis.
Collapse
Affiliation(s)
- Peter K Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, The Institute for Medical Research at North Shore/LIJ, Manhasset, NY 11030, USA.
| |
Collapse
|
44
|
Criswell LA, Pfeiffer KA, Lum RF, Gonzales B, Novitzke J, Kern M, Moser KL, Begovich AB, Carlton VEH, Li W, Lee AT, Ortmann W, Behrens TW, Gregersen PK. Analysis of families in the multiple autoimmune disease genetics consortium (MADGC) collection: the PTPN22 620W allele associates with multiple autoimmune phenotypes. Am J Hum Genet 2005; 76:561-71. [PMID: 15719322 PMCID: PMC1199294 DOI: 10.1086/429096] [Citation(s) in RCA: 445] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2004] [Accepted: 01/20/2005] [Indexed: 12/13/2022] Open
Abstract
Autoimmune disorders constitute a diverse group of phenotypes with overlapping features and a tendency toward familial aggregation. It is likely that common underlying genes are involved in these disorders. Until very recently, no specific alleles--aside from a few common human leukocyte antigen class II genes--had been identified that clearly associate with multiple different autoimmune diseases. In this study, we describe a unique collection of 265 multiplex families assembled by the Multiple Autoimmune Disease Genetics Consortium (MADGC). At least two of nine "core" autoimmune diseases are present in each of these families. These core diseases include rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), multiple sclerosis (MS), autoimmune thyroid disease (Hashimoto thyroiditis or Graves disease), juvenile RA, inflammatory bowel disease (Crohn disease or ulcerative colitis), psoriasis, and primary Sjogren syndrome. We report that a recently described functional single-nucleotide polymorphism (rs2476601, encoding R620W) in the intracellular tyrosine phosphatase (PTPN22) confers risk of four separate autoimmune phenotypes in these families: T1D, RA, SLE, and Hashimoto thyroiditis. MS did not show association with the PTPN22 risk allele. These findings suggest a common underlying etiologic pathway for some, but not all, autoimmune disorders, and they suggest that MS may have a pathogenesis that is distinct from RA, SLE, and T1D. DNA and clinical data for the MADGC families are available to the scientific community; these data will provide a valuable resource for the dissection of the complex genetic factors that underlie the various autoimmune phenotypes.
Collapse
Affiliation(s)
- Lindsey A. Criswell
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Kirsten A. Pfeiffer
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Raymond F. Lum
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Bonnie Gonzales
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Jill Novitzke
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Marlena Kern
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Kathy L. Moser
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Ann B. Begovich
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Victoria E. H. Carlton
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Wentian Li
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Annette T. Lee
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Ward Ortmann
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Timothy W. Behrens
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| | - Peter K. Gregersen
- Rosalind Russell Medical Research Center for Arthritis, Department of Medicine, University of California San Francisco, San Francisco; Robert S. Boas Center for Genomics and Human Genetics, North Shore Long Island Jewish Research Institute, Manhasset, NY; Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis; and Celera Diagnostics, Alameda, CA
| |
Collapse
|
45
|
Begovich AB, Caillier SJ, Alexander HC, Penko JM, Hauser SL, Barcellos LF, Oksenberg JR. The R620W polymorphism of the protein tyrosine phosphatase PTPN22 is not associated with multiple sclerosis. Am J Hum Genet 2005; 76:184-7. [PMID: 15580548 PMCID: PMC1196423 DOI: 10.1086/427244] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Ann B. Begovich
- Celera Diagnostics, Alameda, CA; Department of Neurology, University of California–San Francisco, San Francisco; and Division of Epidemiology, School of Public Health, University of California–Berkeley, Berkeley
| | - Stacy J. Caillier
- Celera Diagnostics, Alameda, CA; Department of Neurology, University of California–San Francisco, San Francisco; and Division of Epidemiology, School of Public Health, University of California–Berkeley, Berkeley
| | - Heather C. Alexander
- Celera Diagnostics, Alameda, CA; Department of Neurology, University of California–San Francisco, San Francisco; and Division of Epidemiology, School of Public Health, University of California–Berkeley, Berkeley
| | - Joanne M. Penko
- Celera Diagnostics, Alameda, CA; Department of Neurology, University of California–San Francisco, San Francisco; and Division of Epidemiology, School of Public Health, University of California–Berkeley, Berkeley
| | - Stephen L. Hauser
- Celera Diagnostics, Alameda, CA; Department of Neurology, University of California–San Francisco, San Francisco; and Division of Epidemiology, School of Public Health, University of California–Berkeley, Berkeley
| | - Lisa F. Barcellos
- Celera Diagnostics, Alameda, CA; Department of Neurology, University of California–San Francisco, San Francisco; and Division of Epidemiology, School of Public Health, University of California–Berkeley, Berkeley
| | - Jorge R. Oksenberg
- Celera Diagnostics, Alameda, CA; Department of Neurology, University of California–San Francisco, San Francisco; and Division of Epidemiology, School of Public Health, University of California–Berkeley, Berkeley
| |
Collapse
|
46
|
Dutta K, Shi H, Cruz-Chu ER, Kami K, Ghose R. Dynamic influences on a high-affinity, high-specificity interaction involving the C-terminal SH3 domain of p67phox. Biochemistry 2004; 43:8094-106. [PMID: 15209505 DOI: 10.1021/bi030268d] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An analysis of the backbone dynamics of the C-terminal Src homology 3 (SH3) domain of p67(phox), p67(phox)SH3(C), in complex with a 32-residue high-affinity (K(d) = 24 nM) peptide, Pf, from the C-terminal region of p47(phox) is presented. This paper represents the first detailed analysis of the backbone dynamics and the ligand-induced changes therein of a high-affinity, high-specificity interaction involving an SH3 domain. The dynamic features are compared with those in the high-affinity, highly specific interaction between the SH3 domain of C-terminal Src kinase (Csk-SH3) and a proline-rich peptide from proline-enriched phosphatase (PEP). Both systems share common dynamic features especially in the canonical PxxP motif recognition surface where slow micro- to millisecond time scale dynamics persist on complex formation especially in several residues that are implicated in ligand recognition and in stabilizing the SH3 fold. These residues are highly conserved in SH3 domains. Ile505, which lies outside the PxxP recognition motif on p67(phox)SH3(C) and is key in conferring high specificity to the p67(phox)SH3(C)/Pf interaction, becomes more disordered upon complex formation. This behavior is similar to that seen in the residues that constitute the specificity surface in Csk-SH3.
Collapse
Affiliation(s)
- Kaushik Dutta
- The New York Structural Biology Center, New York, New York 10031, USA
| | | | | | | | | |
Collapse
|
47
|
Begovich AB, Carlton VEH, Honigberg LA, Schrodi SJ, Chokkalingam AP, Alexander HC, Ardlie KG, Huang Q, Smith AM, Spoerke JM, Conn MT, Chang M, Chang SYP, Saiki RK, Catanese JJ, Leong DU, Garcia VE, McAllister LB, Jeffery DA, Lee AT, Batliwalla F, Remmers E, Criswell LA, Seldin MF, Kastner DL, Amos CI, Sninsky JJ, Gregersen PK. A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis. Am J Hum Genet 2004; 75:330-7. [PMID: 15208781 PMCID: PMC1216068 DOI: 10.1086/422827] [Citation(s) in RCA: 1080] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2004] [Accepted: 05/25/2004] [Indexed: 12/17/2022] Open
Abstract
Rheumatoid arthritis (RA) is the most common systemic autoimmune disease, affecting approximately 1% of the adult population worldwide, with an estimated heritability of 60%. To identify genes involved in RA susceptibility, we investigated the association between putative functional single-nucleotide polymorphisms (SNPs) and RA among white individuals by use of a case-control study design; a second sample was tested for replication. Here we report the association of RA susceptibility with the minor allele of a missense SNP in PTPN22 (discovery-study allelic P=6.6 x 10(-4); replication-study allelic P=5.6 x 10(-8)), which encodes a hematopoietic-specific protein tyrosine phosphatase also known as "Lyp." We show that the risk allele, which is present in approximately 17% of white individuals from the general population and in approximately 28% of white individuals with RA, disrupts the P1 proline-rich motif that is important for interaction with Csk, potentially altering these proteins' normal function as negative regulators of T-cell activation. The minor allele of this SNP recently was implicated in type 1 diabetes, suggesting that the variant phosphatase may increase overall reactivity of the immune system and may heighten an individual carrier's risk for autoimmune disease.
Collapse
|
48
|
Tsutsumi R, Higashi H, Higuchi M, Okada M, Hatakeyama M. Attenuation of Helicobacter pylori CagA x SHP-2 signaling by interaction between CagA and C-terminal Src kinase. J Biol Chem 2003; 278:3664-70. [PMID: 12446738 DOI: 10.1074/jbc.m208155200] [Citation(s) in RCA: 232] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a causative agent of gastric diseases ranging from gastritis to cancer. The CagA protein is the product of the cagA gene carried among virulent H. pylori strains and is associated with severe disease outcomes, most notably gastric carcinoma. CagA is injected from the attached H. pylori into gastric epithelial cells and undergoes tyrosine phosphorylation. The phosphorylated CagA binds and activates SHP-2 phosphatase and thereby induces a growth factor-like morphological change termed the "hummingbird phenotype." In this work, we demonstrate that CagA is also capable of interacting with C-terminal Src kinase (Csk). As is the case with SHP-2, Csk selectively binds tyrosine-phosphorylated CagA via its SH2 domain. Upon complex formation, CagA stimulates Csk, which in turn inactivates the Src family of protein-tyrosine kinases. Because Src family kinases are responsible for CagA phosphorylation, an essential prerequisite of CagA.SHP-2 complex formation and subsequent induction of the hummingbird phenotype, our results indicate that CagA-Csk interaction down-regulates CagA.SHP-2 signaling by both competitively inhibiting CagA.SHP-2 complex formation and reducing levels of CagA phosphorylation. We further demonstrate that CagA.SHP-2 signaling eventually induces apoptosis in AGS cells. Our results thus indicate that CagA-Csk interaction prevents excess cell damage caused by deregulated activation of SHP-2. Attenuation of CagA activity by Csk may enable cagA-positive H. pylori to persistently infect the human stomach for decades while avoiding excess CagA toxicity to the host.
Collapse
Affiliation(s)
- Ryouhei Tsutsumi
- Division of Molecular Oncology, Institute for Genetic Medicine, and Graduate School of Science, Hokkaido University, Sapporo 060-0185, Japan
| | | | | | | | | |
Collapse
|
49
|
Zhu DM, Tibbles HE, Vassilev AO, Uckun FM. SYK and LYN mediate B-cell receptor-independent calcium-induced apoptosis in DT-40 lymphoma B-cells. Leuk Lymphoma 2002; 43:2165-70. [PMID: 12533043 DOI: 10.1080/1042819021000032935] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Here, we report that the calcium ionophore ionomycin induces a massive Ca2+-dependent apoptosis in wildtype DT-40 chicken B lymphoma cells, as well as in BTK-deficient, PLCgamma2-deficient and IP3 receptor-deficient DT-40 cells, but not in LYN- or SYK-deficient DT-40 cells. Notably, the deficiency of CSK, a negative regulator of Src-family PTK, promoted ionomycin-induced apoptosis of DT-40 cells. Reconstitution of SYK-deficient cells with wild-type SYK restored the apoptotic response of the cells to ionomycin, but the expression of FYN or LCK in LYN-deficient cells did not restore the apoptotic response of LYN-deficient cells. Taken together, our data suggests that both LYN and SYK, but not BTK, FYN or LCK, are crucial mediators of BCR-independent Ca2+-induced apoptosis in DT-40 lymphoma B cells.
Collapse
Affiliation(s)
- De-Min Zhu
- Department of Immunology, Parker Hughes Cancer Center Parker Hughes Institute, 2669 Patton Road, Roseville, St. Paul, MN 55113, USA
| | | | | | | |
Collapse
|
50
|
Kami K, Takeya R, Sumimoto H, Kohda D. Diverse recognition of non-PxxP peptide ligands by the SH3 domains from p67(phox), Grb2 and Pex13p. EMBO J 2002; 21:4268-76. [PMID: 12169629 PMCID: PMC126167 DOI: 10.1093/emboj/cdf428] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The basic function of the Src homology 3 (SH3) domain is considered to be binding to proline-rich sequences containing a PxxP motif. Recently, many SH3 domains, including those from Grb2 and Pex13p, were reported to bind sequences lacking a PxxP motif. We report here that the 22 residue peptide lacking a PxxP motif, derived from p47(phox), binds to the C-terminal SH3 domain from p67(phox). We applied the NMR cross-saturation method to locate the interaction sites for the non-PxxP peptides on their cognate SH3 domains from p67(phox), Grb2 and Pex13p. The binding site of the Grb2 SH3 partially overlapped the conventional PxxP-binding site, whereas those of p67(phox) and Pex13p SH3s are located in different surface regions. The non-PxxP peptide from p47(phox) binds to the p67(phox) SH3 more tightly when it extends to the N-terminus to include a typical PxxP motif, which enabled the structure determination of the complex, to reveal that the non-PxxP peptide segment interacted with the p67(phox) SH3 in a compact helix-turn-helix structure (PDB entry 1K4U).
Collapse
Affiliation(s)
- Keiichiro Kami
- Department of Structural Biology, Biomolecular Engineering Research Institute, 6-2-3, Furuedai, Suita, Osaka 565-0874 and Department of Biochemistry and Molecular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Present address: Department of Biochemistry and Molecular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Present address: Department of Structural Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Corresponding author e-mail:
| | - Ryu Takeya
- Department of Structural Biology, Biomolecular Engineering Research Institute, 6-2-3, Furuedai, Suita, Osaka 565-0874 and Department of Biochemistry and Molecular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Present address: Department of Biochemistry and Molecular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Present address: Department of Structural Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Corresponding author e-mail:
| | - Hideki Sumimoto
- Department of Structural Biology, Biomolecular Engineering Research Institute, 6-2-3, Furuedai, Suita, Osaka 565-0874 and Department of Biochemistry and Molecular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Present address: Department of Biochemistry and Molecular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Present address: Department of Structural Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Corresponding author e-mail:
| | - Daisuke Kohda
- Department of Structural Biology, Biomolecular Engineering Research Institute, 6-2-3, Furuedai, Suita, Osaka 565-0874 and Department of Biochemistry and Molecular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Present address: Department of Biochemistry and Molecular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Present address: Department of Structural Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan Corresponding author e-mail:
| |
Collapse
|