1
|
Wu CS, Liu FC, Lin SC, Chyuan IT. Regulation of T cell receptor (TCR) signaling by tyrosine phosphatases: Recent advances and implication for therapeutic approach in autoimmune diseases. J Formos Med Assoc 2025:S0929-6646(25)00192-5. [PMID: 40287371 DOI: 10.1016/j.jfma.2025.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
The effector function of T cells is critical for regulation of the initiation and progression of autoimmune diseases; whereas the T cell activation and homeostasis are tightly controlled by signals from T cell receptor (TCR). The early TCR signaling pathways are dependent on rapid phosphorylation and dephosphorylation of multiple signaling proteins in the TCR complex. These processes are tightly regulated by the interplay between protein kinases and phosphatases, leading to T cell activation. Genetic polymorphisms of these kinases or phosphatases have been linked to an increased susceptibility to autoimmune disorders in humans. Mice with deficiencies in these corresponding genes often exhibit T cell hyper-reactivity and autoimmune phenotypes in animal models. Tyrosine phosphatases have been demonstrated to alter T cell fate by negatively regulating early TCR signaling. Therefore, the tyrosine phosphatases that regulate TCR signaling are emerging as potential therapeutic targets to modulate T cell responses for the treatment of autoimmune diseases. In this review, we provide an overview of the current progress and perspectives of tyrosine phosphatases that regulate TCR signaling in T cell activation, and their potential as therapeutic targets for autoimmune diseases.
Collapse
Affiliation(s)
- Chien-Sheng Wu
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, 22000, Taiwan
| | - Feng-Cheng Liu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Shih-Chang Lin
- Department of Internal Medicine, Cathay General Hospital, Taipei, 10630, Taiwan; School of Medicine, Fu-Jen Catholic University, New Taipei City, 242062, Taiwan
| | - I-Tsu Chyuan
- Department of Internal Medicine, Cathay General Hospital, Taipei, 10630, Taiwan; School of Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Research, Cathay General Hospital, Taipei, 10630, Taiwan.
| |
Collapse
|
2
|
Bennett AM, Tiganis T. Protein Tyrosine Phosphatases in Metabolism: A New Frontier for Therapeutics. Annu Rev Physiol 2025; 87:301-324. [PMID: 39531392 DOI: 10.1146/annurev-physiol-022724-105540] [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] [Indexed: 11/16/2024]
Abstract
The increased prevalence of chronic metabolic disorders, including obesity and type 2 diabetes and their associated comorbidities, are among the world's greatest health and economic challenges. Metabolic homeostasis involves a complex interplay between hormones that act on different tissues to elicit changes in the storage and utilization of energy. Such processes are mediated by tyrosine phosphorylation-dependent signaling, which is coordinated by the opposing actions of protein tyrosine kinases and protein tyrosine phosphatases (PTPs). Perturbations in the functions of PTPs can be instrumental in the pathophysiology of metabolic diseases. The goal of this review is to highlight key advances in our understanding of how PTPs control body weight and glucose metabolism, as well as their contributions to obesity and type 2 diabetes. The emerging appreciation of the integrated functions of PTPs in metabolism, coupled with significant advances in pharmaceutical strategies aimed at targeting this class of enzymes, marks the advent of a new frontier in combating metabolic disorders.
Collapse
Affiliation(s)
- Anton M Bennett
- Yale Center for Molecular and Systems Metabolism, New Haven, Connecticut, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia;
| |
Collapse
|
3
|
Li Q, Tian P, Guo M, Liu X, Su T, Tang M, Meng B, Yu L, Yang Y, Liu Y, Li Y, Li J. Spermidine Associated with Gut Microbiota Protects Against MRSA Bloodstream Infection by Promoting Macrophage M2 Polarization. ACS Infect Dis 2024; 10:3751-3764. [PMID: 39382005 PMCID: PMC11559170 DOI: 10.1021/acsinfecdis.3c00669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 09/01/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a major human pathogen that causes various diseases. Extensive researches highlight the significant role of gut microbiota and its metabolites, particularly spermidine, in infectious diseases. However, the immunomodulatory mechanisms of spermidine in MRSA-induced bloodstream infection remain unclear. Here, we confirmed the protective effects of spermidine in bloodstream infection in mice. Spermidine reduced the bacterial load and expression of inflammatory factors by shifting the macrophage phenotype to an anti-inflammatory phenotype, ultimately prolonging the survival of the infected mice. The protective effect against MRSA infection may rely on the elevated expression of protein tyrosine phosphatase nonreceptor 2 (PTPN2). Collectively, these findings confirm the immunoprotective effects of spermidine via binding to PTPN2 in MRSA bloodstream infection, providing new ideas for the treatment of related infectious diseases.
Collapse
Affiliation(s)
- Qingqing Li
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Infectious Diseases
& Institute of Bacterial Resistance, Anhui Medical
University, Hefei 230022, China
| | - Ping Tian
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Infectious Diseases
& Institute of Bacterial Resistance, Anhui Medical
University, Hefei 230022, China
| | - Mingjuan Guo
- Department of Hepatology, The First
Affiliated Hospital of Jilin University, Changchun 130021,
China
| | - Xiaoqiang Liu
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
| | - Tingting Su
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
| | - Mingyang Tang
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Infectious Diseases
& Institute of Bacterial Resistance, Anhui Medical
University, Hefei 230022, China
| | - Bao Meng
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Infectious Diseases
& Institute of Bacterial Resistance, Anhui Medical
University, Hefei 230022, China
| | - Liang Yu
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Infectious Diseases
& Institute of Bacterial Resistance, Anhui Medical
University, Hefei 230022, China
| | - Yi Yang
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Infectious Diseases
& Institute of Bacterial Resistance, Anhui Medical
University, Hefei 230022, China
| | - Yanyan Liu
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Infectious Diseases
& Institute of Bacterial Resistance, Anhui Medical
University, Hefei 230022, China
| | - Yasheng Li
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Infectious Diseases
& Institute of Bacterial Resistance, Anhui Medical
University, Hefei 230022, China
| | - Jiabin Li
- Department of Infectious Diseases & Anhui Center
for Surveillance of Bacterial Resistance, The First Affiliated Hospital of
Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Infectious Diseases
& Institute of Bacterial Resistance, Anhui Medical
University, Hefei 230022, China
| |
Collapse
|
4
|
Willis TW, Gkrania-Klotsas E, Wareham NJ, McKinney EF, Lyons PA, Smith KGC, Wallace C. Leveraging pleiotropy identifies common-variant associations with selective IgA deficiency. Clin Immunol 2024; 268:110356. [PMID: 39241920 DOI: 10.1016/j.clim.2024.110356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/22/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Selective IgA deficiency (SIgAD) is the most common inborn error of immunity (IEI). Unlike many IEIs, evidence of a role for highly penetrant rare variants in SIgAD is lacking. Previous SIgAD studies have had limited power to identify common variants due to their small sample size. We overcame this problem first through meta-analysis of two existing GWAS. This identified four novel common-variant associations and enrichment of SIgAD-associated variants in genes linked to Mendelian IEIs. SIgAD showed evidence of shared genetic architecture with serum IgA and a number of immune-mediated diseases. We leveraged this pleiotropy through the conditional false discovery rate procedure, conditioning our SIgAD meta-analysis on large GWAS of asthma and rheumatoid arthritis, and our own meta-analysis of serum IgA. This identified an additional 18 variants, increasing the number of known SIgAD-associated variants to 27 and strengthening the evidence for a polygenic, common-variant aetiology for SIgAD.
Collapse
Affiliation(s)
- Thomas W Willis
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK; Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK.
| | - Effrossyni Gkrania-Klotsas
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK; Department of Infectious Diseases, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
| | - Nicholas J Wareham
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Eoin F McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kenneth G C Smith
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Chris Wallace
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK; Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Li HY, Jing YM, Shen X, Tang MY, Shen HH, Li XW, Wang ZS, Su F. Protein tyrosine phosphatase non-receptor II: A possible biomarker of poor prognosis and mediator of immune evasion in hepatocellular carcinoma. World J Gastrointest Oncol 2024; 16:3913-3931. [PMID: 39350977 PMCID: PMC11438766 DOI: 10.4251/wjgo.v16.i9.3913] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/03/2024] [Accepted: 07/08/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND The incidence of primary liver cancer is increasing year by year. In 2022 alone, more than 900000 people were diagnosed with liver cancer worldwide, with hepatocellular carcinoma (HCC) accounting for 75%-85% of cases. HCC is the most common primary liver cancer. China has the highest incidence and mortality rate of HCC in the world, and it is one of the malignant tumors that seriously threaten the health of Chinese people. The onset of liver cancer is occult, the early cases lack typical clinical symptoms, and most of the patients are already in the middle and late stage when diagnosed. Therefore, it is very important to find new markers for the early detection and diagnosis of liver cancer, improve the therapeutic effect, and improve the prognosis of patients. Protein tyrosine phosphatase non-receptor 2 (PTPN2) has been shown to be associated with colorectal cancer, triple-negative breast cancer, non-small cell lung cancer, and prostate cancer, but its biological role and function in tumors remain to be further studied. AIM To combine the results of relevant data obtained from The Cancer Genome Atlas (TCGA) to provide the first in-depth analysis of the biological role of PTPN2 in HCC. METHODS The expression of PTPN2 in HCC was first analyzed based on the TCGA database, and the findings were then verified by immunohistochemical staining, quantitative real-time polymerase chain reaction (qRT-PCR), and immunoblotting. The value of PTPN2 in predicting the survival of patients with HCC was assessed by analyzing the relationship between PTPN2 expression in HCC tissues and clinicopathological features. Finally, the potential of PTPN2 affecting immune escape of liver cancer was evaluated by tumor immune dysfunction and exclusion and immunohistochemical staining. RESULTS The results of immunohistochemical staining, qRT-PCR, and immunoblotting in combination with TCGA database analysis showed that PTPN2 was highly expressed and associated with a poor prognosis in HCC patients. Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed that PTPN2 was associated with various pathways, including cancer-related pathways, the Notch signaling pathway, and the MAPK signaling pathway. Gene Set Enrichment Analysis showed that PTPN2 was highly expressed in various immune-related pathways, such as the epithelial mesenchymal transition process. A risk model score based on PTPN2 showed that immune escape was significantly enhanced in the high-risk group compared with the low-risk group. CONCLUSION This study investigated PTPN2 from multiple biological perspectives, revealing that PTPN2 can function as a biomarker of poor prognosis and mediate immune evasion in HCC.
Collapse
Affiliation(s)
- Hui-Yuan Li
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Yi-Ming Jing
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Xue Shen
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Ming-Yue Tang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Hong-Hong Shen
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Xin-Wei Li
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Zi-Shu Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Fang Su
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, Anhui Province, China
| |
Collapse
|
6
|
Jeanpierre M, Cognard J, Tusseau M, Riller Q, Bui LC, Berthelet J, Laurent A, Crickx E, Parlato M, Stolzenberg MC, Suarez F, Leverger G, Aladjidi N, Collardeau-Frachon S, Pietrement C, Malphettes M, Froissart A, Bole-Feysot C, Cagnard N, Rodrigues Lima F, Walzer T, Rieux-Laucat F, Belot A, Mathieu AL. Haploinsufficiency in PTPN2 leads to early-onset systemic autoimmunity from Evans syndrome to lupus. J Exp Med 2024; 221:e20232337. [PMID: 39028869 PMCID: PMC11259789 DOI: 10.1084/jem.20232337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/17/2024] [Accepted: 06/26/2024] [Indexed: 07/21/2024] Open
Abstract
An exome sequencing strategy employed to identify pathogenic variants in patients with pediatric-onset systemic lupus or Evans syndrome resulted in the discovery of six novel monoallelic mutations in PTPN2. PTPN2 is a phosphatase that acts as an essential negative regulator of the JAK/STAT pathways. All mutations led to a loss of PTPN2 regulatory function as evidenced by in vitro assays and by hyperproliferation of patients' T cells. Furthermore, patients exhibited high serum levels of inflammatory cytokines, mimicking the profile observed in individuals with gain-of-function mutations in STAT factors. Flow cytometry analysis of patients' blood cells revealed typical alterations associated with autoimmunity and all patients presented with autoantibodies. These findings further supported the notion that a loss of function in negative regulators of cytokine pathways can lead to a broad spectrum of autoimmune manifestations and that PTPN2 along with SOCS1 haploinsufficiency constitute a new group of monogenic autoimmune diseases that can benefit from targeted therapy.
Collapse
Affiliation(s)
- Marie Jeanpierre
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Jade Cognard
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
| | - Maud Tusseau
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
- Department of Medical Genetics, Hospices Civils de Lyon, Bron, France
| | - Quentin Riller
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Linh-Chi Bui
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Jérémy Berthelet
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, Paris, France
| | - Audrey Laurent
- National Referee Centre for Pediatric-Onset Rheumatism and Autoimmune Diseases, Hospices Civils de Lyon, Pediatric Nephrology, Rheumatology, Dermatology Unit, Mother and Children University Hospital; Lyon, France
| | - Etienne Crickx
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
- Service de Médecine Interne, Centre National de Référence des Cytopénies Auto-immunes de L’adulte, Hôpital Henri Mondor, Fédération Hospitalo-Universitaire TRUE InnovaTive TheRapy for ImmUne disordErs, Assistance Publique Hôpitaux de Paris, Université Paris Est Créteil, Créteil, France
| | - Marianna Parlato
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Marie-Claude Stolzenberg
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Felipe Suarez
- Department of Adult Hematology, Necker-Enfants Malades University Hospital and Centre de Référence des déficits Immunitaires Héréditaires, Assistance Publique Hôpitaux de Paris, INSERM U1163, Imagine Institute, Université Paris Cité, Paris, France
| | - Guy Leverger
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, UMR_S938, Assistance Publique Hôpitaux de Paris, Groupe Hospitalier Sorbonne Université, Hôpital Armand Trousseau, Paris, France
| | - Nathalie Aladjidi
- Centre de Référence National des Cytopénies Auto-immunes de l’Enfant, Bordeaux, France
- Pediatric Oncology Hemato-Immunology Unit, University Hospital, Plurithématique Centre d’Investigation Clinique, 1401, INSERM, Bordeaux, France
| | - Sophie Collardeau-Frachon
- Institute of Pathology, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Société Française de Foetopathologie Paris, Paris, France
| | - Christine Pietrement
- Centre Hospitalier Universitaire de Reims, Service de Pédiatrie Spécialisée et Généralisée, Université Reims Champagne Ardenne, Reims, France
| | - Marion Malphettes
- Service d’Immunopathologie Clinique, Saint Louis Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Antoine Froissart
- Service Médecine Interne, Hôpital Intercommunal de Créteil, Créteil, France
| | - Christine Bole-Feysot
- Genomic Platform, INSERM UMR 1163, Imagine Institute, University Paris Cité, Paris, France
| | - Nicolas Cagnard
- Bioinformatic Platform, INSERM UMR 1163, Imagine Institute, University Paris Cité, Paris, France
| | | | - Thierry Walzer
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
| | - Frédéric Rieux-Laucat
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute, Université Paris Cité, INSERM UMR 1163, Paris, France, IHU-Imagine, Université de Paris, Paris, France
| | - Alexandre Belot
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
- National Referee Centre for Pediatric-Onset Rheumatism and Autoimmune Diseases, Hospices Civils de Lyon, Pediatric Nephrology, Rheumatology, Dermatology Unit, Mother and Children University Hospital; Lyon, France
| | - Anne-Laure Mathieu
- Centre International de Recherche en Infectiologie, Inserm, U1111, CNRS, UMR5308, École Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
7
|
McBride DA, Wang JS, Johnson WT, Bottini N, Shah NJ. ABCD of IA: A multi-scale agent-based model of T cell activation in inflammatory arthritis. Biomater Sci 2024; 12:2041-2056. [PMID: 38349277 PMCID: PMC11757005 DOI: 10.1039/d3bm01674a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Biomaterial-based agents have been demonstrated to regulate the function of immune cells in models of autoimmunity. However, the complexity of the kinetics of immune cell activation can present a challenge in optimizing the dose and frequency of administration. Here, we report a model of autoreactive T cell activation, which are key drivers in autoimmune inflammatory joint disease. The model is termed a multi-scale Agent-Based, Cell-Driven model of Inflammatory Arthritis (ABCD of IA). Using kinetic rate equations and statistical theory, ABCD of IA simulated the activation and presentation of autoantigens by dendritic cells, interactions with cognate T cells and subsequent T cell proliferation in the lymph node and IA-affected joints. The results, validated with in vivo data from the T cell driven SKG mouse model, showed that T cell proliferation strongly correlated with the T cell receptor (TCR) affinity distribution (TCR-ad), with a clear transition state from homeostasis to an inflammatory state. T cell proliferation was strongly dependent on the amount of antigen in antigenic stimulus event (ASE) at low concentrations. On the other hand, inflammation driven by Th17-inducing cytokine mediated T cell phenotype commitment was influenced by the initial level of Th17-inducing cytokines independent of the amount of arthritogenic antigen. The introduction of inhibitory artificial antigen presenting cells (iaAPCs), which locally suppress T cell activation, reduced T cell proliferation in a dose-dependent manner. The findings in this work set up a framework based on theory and modeling to simulate personalized therapeutic strategies in IA.
Collapse
Affiliation(s)
- David A McBride
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.
| | - James S Wang
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Wade T Johnson
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Nunzio Bottini
- Kao Autoimmunity Institute and Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nisarg J Shah
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
8
|
Liang S, Tran E, Du X, Dong J, Sudholz H, Chen H, Qu Z, Huntington ND, Babon JJ, Kershaw NJ, Zhang ZY, Baell JB, Wiede F, Tiganis T. A small molecule inhibitor of PTP1B and PTPN2 enhances T cell anti-tumor immunity. Nat Commun 2023; 14:4524. [PMID: 37500611 PMCID: PMC10374545 DOI: 10.1038/s41467-023-40170-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/15/2023] [Indexed: 07/29/2023] Open
Abstract
The inhibition of protein tyrosine phosphatases 1B (PTP1B) and N2 (PTPN2) has emerged as an exciting approach for bolstering T cell anti-tumor immunity. ABBV-CLS-484 is a PTP1B/PTPN2 inhibitor in clinical trials for solid tumors. Here we have explored the therapeutic potential of a related small-molecule-inhibitor, Compound-182. We demonstrate that Compound-182 is a highly potent and selective active site competitive inhibitor of PTP1B and PTPN2 that enhances T cell recruitment and activation and represses the growth of tumors in mice, without promoting overt immune-related toxicities. The enhanced anti-tumor immunity in immunogenic tumors can be ascribed to the inhibition of PTP1B/PTPN2 in T cells, whereas in cold tumors, Compound-182 elicited direct effects on both tumor cells and T cells. Importantly, treatment with Compound-182 rendered otherwise resistant tumors sensitive to α-PD-1 therapy. Our findings establish the potential for small molecule inhibitors of PTP1B and PTPN2 to enhance anti-tumor immunity and combat cancer.
Collapse
Affiliation(s)
- Shuwei Liang
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Eric Tran
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Xin Du
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Jiajun Dong
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Harrison Sudholz
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Hao Chen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, 3052, Australia
| | - Zihan Qu
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Nicholas D Huntington
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Jeffrey J Babon
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, 3052, Australia
| | - Nadia J Kershaw
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, 3052, Australia
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Jonathan B Baell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Lyterian Therapeutics, South San Francisco, San Francisco, CA, 94080, USA
| | - Florian Wiede
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
9
|
Liang S, Tran E, Du X, Dong J, Sudholz H, Chen H, Qu Z, Huntington N, Babon J, Kershaw N, Zhang ZY, Baell J, Wiede F, Tiganis T. A small molecule inhibitor of PTP1B and PTPN2 enhances T cell anti-tumor immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545220. [PMID: 37397992 PMCID: PMC10312756 DOI: 10.1101/2023.06.16.545220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The inhibition of protein tyrosine phosphatases (PTPs), such as PTP1B and PTPN2 that function as intracellular checkpoints, has emerged as an exciting new approach for bolstering T cell anti-tumor immunity to combat cancer. ABBV-CLS-484 is a dual PTP1B and PTPN2 inhibitor currently in clinical trials for solid tumors. Here we have explored the therapeutic potential of targeting PTP1B and PTPN2 with a related small molecule inhibitor, Compound 182. We demonstrate that Compound 182 is a highly potent and selective active site competitive inhibitor of PTP1B and PTPN2 that enhances antigen-induced T cell activation and expansion ex vivo and represses the growth of syngeneic tumors in C57BL/6 mice without promoting overt immune-related toxicities. Compound 182 repressed the growth of immunogenic MC38 colorectal and AT3-OVA mammary tumors as well as immunologically cold AT3 mammary tumors that are largely devoid of T cells. Treatment with Compound 182 increased both the infiltration and activation of T cells, as well as the recruitment of NK cells and B cells that promote anti-tumor immunity. The enhanced anti-tumor immunity in immunogenic AT3-OVA tumors could be ascribed largely to the inhibition of PTP1B/PTPN2 in T cells, whereas in cold AT3 tumors, Compound 182 elicited both direct effects on tumor cells and T cells to facilitate T cell recruitment and thereon activation. Importantly, treatment with Compound 182 rendered otherwise resistant AT3 tumors sensitive to anti-PD1 therapy. Our findings establish the potential for small molecule active site inhibitors of PTP1B and PTPN2 to enhance anti-tumor immunity and combat cancer.
Collapse
|
10
|
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
|
11
|
Rodrigo S, Senasinghe K, Quazi S. Molecular and therapeutic effect of CRISPR in treating cancer. Med Oncol 2023; 40:81. [PMID: 36650384 PMCID: PMC9845174 DOI: 10.1007/s12032-022-01930-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/13/2022] [Indexed: 01/18/2023]
Abstract
Cancer has become one of the common causes of mortality around the globe due to mutations in the genome which allows rapid growth of cells uncontrollably without repairing DNA errors. Cancers could arise due alterations in DNA repair mechanisms (errors in mismatch repair genes), activation of oncogenes and inactivation of tumor suppressor genes. Each cancer type is different and each individual has a unique genetic change which leads them to cancer. Studying genetic and epigenetic alterations in the genome leads to understanding the underlying features. CAR T therapy over other immunotherapies such as monoclonal antibodies, immune checkpoint inhibitors, cancer vaccines and adoptive cell therapies has been widely used to treat cancer in recent days and gene editing has now become one of the promising treatments for many genetic diseases. This tool allows scientists to change the genome by adding, removing or altering genetic material of an organism. Due to advance in genetics and novel molecular techniques such as CRISPR, TALEN these genes can be edited in such a way that their original function could be replaced which in turn improved the treatment possibilities and can be used against malignancies and even cure cancer in future along with CAR T cell therapy due to the specific recognition and attacking of tumor.
Collapse
Affiliation(s)
- Sawani Rodrigo
- Human Genetics Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Kaveesha Senasinghe
- Human Genetics Unit, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - Sameer Quazi
- GenLab Biosolutions Private Limited, Bengaluru, Karnataka, 560043, India.
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge, UK.
- School of Health Sciences, The University of Manchester, Manchester, UK.
- SCAMT Institute, ITMO University, St. Petersburg, Russia.
| |
Collapse
|
12
|
Zhu Z, Tang R, Huff S, Kummetha IR, Wang L, Li N, Rana TM. Small-molecule PTPN2 Inhibitors Sensitize Resistant Melanoma to Anti-PD-1 Immunotherapy. CANCER RESEARCH COMMUNICATIONS 2023; 3:119-129. [PMID: 36968224 PMCID: PMC10035454 DOI: 10.1158/2767-9764.crc-21-0186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 08/23/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Although immune checkpoint inhibitors targeting T-cell immunoregulatory proteins have revolutionized cancer treatment, they are effective only in a limited number of patients, and new strategies are needed to enhance tumor responses to immunotherapies. Deletion of protein tyrosine phosphatase non-receptor type 2 (Ptpn2), a regulator of growth factor and cytokine signaling pathways, has been shown to sensitize murine B16F10 melanoma cells to IFNγ and anti-PD-1 immunotherapy. Here, we investigated the potential therapeutic utility of small-molecule PTPN2 inhibitors. Ten inhibitors were synthesized on the basis of in silico modeling and structure-based design and functionally tested in vitro and in vivo. We show that the inhibitors had little effect on B16F10 cells alone, but effectively sensitized the tumor cells to IFNγ treatment in vitro and to anti-PD-1 therapy in vivo. Under both conditions, Ptpn2 inhibitor cotreatment suppressed B16F10 cell growth and enhanced Stat1 phosphorylation and expression of IFNγ response genes. In vivo, PTPN2 inhibitor cotreatment significantly reduced melanoma and colorectal tumor growth and enhanced mouse survival compared with anti-PD-1 treatment alone, and this was accompanied by increased tumor infiltration by granzyme B+ CD8+ T cells. Similar results were obtained with representative murine and human colon cancer and lung cancer cell lines. Collectively, these results demonstrate that small-molecule inhibitors of PTPN2 may have clinical utility as sensitizing agents for immunotherapy-resistant cancers. Significance To enhance the effectiveness of immunotherapies in resistant or nonresponsive cancers, it is important to develop inhibitors of enzymes that negatively influence the outcome of treatments. We have designed and evaluated small-molecule inhibitors of PTPN2 demonstrating that these compounds may have clinical utility as sensitizing agents for immunotherapy-resistant cancers.
Collapse
Affiliation(s)
- Zhouting Zhu
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Rachel Tang
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Sarah Huff
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Indrasena Reddy Kummetha
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Lingling Wang
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Na Li
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
| | - Tariq M. Rana
- Division of Genetics, Department of Pediatrics, Program in Immunology, Institute for Genomic Medicine, University of California San Diego, La Jolla, California
- San Diego Center for Precision Immunotherapy, Moores Cancer Center, University of California San Diego, La Jolla, California
| |
Collapse
|
13
|
Cai X, Lymbery AJ, Armstrong NJ, Gao C, Ma L, Li C. Systematic identification and characterization of lncRNAs and lncRNA-miRNA-mRNA networks in the liver of turbot (Scophthalmus maximus L.) induced with Vibrio anguillarum. FISH & SHELLFISH IMMUNOLOGY 2022; 131:21-29. [PMID: 36170960 DOI: 10.1016/j.fsi.2022.09.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/05/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
Long noncoding RNAs (lncRNAs), can regulate mRNA by targeting miRNA in a competing endogenous RNA network, have become a hot topic in the research of fish immune mechanism recent years. While in turbot (Scophthalmus maximus L.), an economically important marine fish, there are limited researches about the role of lncRNAs in its immune response to bacterial infection. In this study, a total of 184 differentially expressed lncRNAs (DElncRNAs) were systematically identified and characterized using whole-transcriptome sequencing of the liver of turbot challenged with Vibrioanguillarum at 0 h (control) and three different time points post infection (2 h, 12 h and 24 h, respectively). Subsequently, GO and KEGG signaling pathways of differentially expressed lncRNAs were analyzed to predict their function. We found that lncRNAs in our results were significantly enriched in several immune-related signaling pathways, including the NOD-like receptor signaling pathway, Toll-like receptor signaling pathway, Cytokine-cytokine receptor, MAPK signaling pathway, phagosome, PPAR signaling pathway and the regulation of autophagy. In addition, a total of 492 DE lncRNA - DE miRNA -DE mRNA networks were identified at three different time points post infection, which were consisted of 102 networks at 2 h, 122 networks at 12 h and 81 networks at 24 h post infection, respectively. Noticeably, 92 of these regulated networks were immune-related. These observations suggested that lncRNAs can regulate the expression of immune-related genes in the response to bacterial infection in turbot. Moreover, our findings would provide a new insight into the immune response of turbot to pathogen infection and lay a foundation for future study.
Collapse
Affiliation(s)
- Xin Cai
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China; Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, School of Veterinary & Life Sciences, Murdoch University, South Street, Murdoch, Western Australia, 6150, Australia
| | - Alan J Lymbery
- Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, School of Veterinary & Life Sciences, Murdoch University, South Street, Murdoch, Western Australia, 6150, Australia
| | - Nicola J Armstrong
- Department of Mathematics and Statistics, Curtin University, Kent Street, Bentley, Perth, WA, 6102, Australia
| | - Chengbin Gao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China; Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, School of Veterinary & Life Sciences, Murdoch University, South Street, Murdoch, Western Australia, 6150, Australia
| | - Le Ma
- Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, School of Veterinary & Life Sciences, Murdoch University, South Street, Murdoch, Western Australia, 6150, Australia
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
14
|
Hocking AM, Buckner JH. Genetic basis of defects in immune tolerance underlying the development of autoimmunity. Front Immunol 2022; 13:972121. [PMID: 35979360 PMCID: PMC9376219 DOI: 10.3389/fimmu.2022.972121] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/14/2022] [Indexed: 12/15/2022] Open
Abstract
Genetic variants associated with susceptibility to autoimmune disease have provided important insight into the mechanisms responsible for the loss of immune tolerance and the subsequent development of autoantibodies, tissue damage, and onset of clinical disease. Here, we review how genetic variants shared across multiple autoimmune diseases have contributed to our understanding of global tolerance failure, focusing on variants in the human leukocyte antigen region, PTPN2 and PTPN22, and their role in antigen presentation and T and B cell homeostasis. Variants unique to a specific autoimmune disease such as those in PADI2 and PADI4 that are associated with rheumatoid arthritis are also discussed, addressing their role in disease-specific immunopathology. Current research continues to focus on determining the functional consequences of autoimmune disease-associated variants but has recently expanded to variants in the non-coding regions of the genome using novel approaches to investigate the impact of these variants on mechanisms regulating gene expression. Lastly, studying genetic risk variants in the setting of autoimmunity has clinical implications, helping predict who will develop autoimmune disease and also identifying potential therapeutic targets.
Collapse
|
15
|
Tang X, Qi C, Zhou H, Liu Y. Critical roles of PTPN family members regulated by non-coding RNAs in tumorigenesis and immunotherapy. Front Oncol 2022; 12:972906. [PMID: 35957898 PMCID: PMC9360549 DOI: 10.3389/fonc.2022.972906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/04/2022] [Indexed: 12/22/2022] Open
Abstract
Since tyrosine phosphorylation is reversible and dynamic in vivo, the phosphorylation state of proteins is controlled by the opposing roles of protein tyrosine kinases (PTKs) and protein tyrosine phosphatase (PTPs), both of which perform critical roles in signal transduction. Of these, intracellular non-receptor PTPs (PTPNs), which belong to the largest class I cysteine PTP family, are essential for the regulation of a variety of biological processes, including but not limited to hematopoiesis, inflammatory response, immune system, and glucose homeostasis. Additionally, a substantial amount of PTPNs have been identified to hold crucial roles in tumorigenesis, progression, metastasis, and drug resistance, and inhibitors of PTPNs have promising applications due to striking efficacy in antitumor therapy. Hence, the aim of this review is to summarize the role played by PTPNs, including PTPN1/PTP1B, PTPN2/TC-PTP, PTPN3/PTP-H1, PTPN4/PTPMEG, PTPN6/SHP-1, PTPN9/PTPMEG2, PTPN11/SHP-2, PTPN12/PTP-PEST, PTPN13/PTPL1, PTPN14/PEZ, PTPN18/PTP-HSCF, PTPN22/LYP, and PTPN23/HD-PTP, in human cancer and immunotherapy and to comprehensively describe the molecular pathways in which they are implicated. Given the specific roles of PTPNs, identifying potential regulators of PTPNs is significant for understanding the mechanisms of antitumor therapy. Consequently, this work also provides a review on the role of non-coding RNAs (ncRNAs) in regulating PTPNs in tumorigenesis and progression, which may help us to find effective therapeutic agents for tumor therapy.
Collapse
Affiliation(s)
- Xiaolong Tang
- Department of Clinical Laboratory Diagnostics, Binzhou Medical University, Binzhou, China
| | - Chumei Qi
- Department of Clinical Laboratory, Dazhou Women and Children’s Hospital, Dazhou, China
| | - Honghong Zhou
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Honghong Zhou, ; Yongshuo Liu,
| | - Yongshuo Liu
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- *Correspondence: Honghong Zhou, ; Yongshuo Liu,
| |
Collapse
|
16
|
Chatterjee K, Dutta AK, Goel A, Aaron R, Balakrishnan V, Thomas A, John A, Jaleel R, David D, Kurien RT, Chowdhury SD, Simon EG, Joseph AJ, Premkumar P, Pulimood AB. Common polymorphisms of protein tyrosine phosphate non-receptor type 2 gene are not associated with risk of Crohn’s disease in Indian. World J Gastrointest Pathophysiol 2022; 13:114-123. [PMID: 36161231 PMCID: PMC9350595 DOI: 10.4291/wjgp.v13.i4.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/18/2022] [Accepted: 05/23/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Multiple genetic risk factors for Crohn’s disease (CD) have been identified. However, these observations are not consistent across different populations. The protein tyrosine phosphate non-receptor type 2 (PTPN2) gene plays a role in various aspects of host defense including epithelial barrier function, autophagy, and innate and adaptive immune response. Two common polymorphisms in the PTPN2 gene (rs2542151 and rs7234029) have been associated with risk of CD in Western countries.
AIM To evaluate the association of PTPN2 gene polymorphisms with risk of CD in Indian population.
METHODS We conducted a prospective case-control study. Patients with CD were recruited, and their clinical and investigation details were noted. Controls were patients without organic gastrointestinal disease or other comorbid illnesses. Two common polymorphisms in the PTPN2 gene (rs2542151 and rs7234029) were assessed. DNA was extracted from peripheral blood samples of cases and controls and target DNA was amplified using specific sets of primers. The amplified fragments were digested with restriction enzymes and the presence of polymorphism was detected by restriction fragment length polymorphism. The frequency of alleles was determined. The frequencies of genotypes and alleles were compared between cases and controls to look for significant differences.
RESULTS A total of 108 patients with CD (mean age 37.5 ± 12.7 years, females 42.6%) and 100 controls (mean age 39.9 ± 13.5 years, females 37%) were recruited. For the single nucleotide polymorphism (SNP) rs7234029, the overall frequency of G variant genotype (AG or GG) was noted to be significantly lower in the cases compared to controls (35.2% vs 50%, P = 0.05). For the SNP rs2542151, the overall frequency of G variant genotype (GT or GG) was noted to be similar in cases compared to controls (43.6% vs 47%, P = 0.73). There were no significant differences in minor allele (G) frequency for both polymorphisms between the cases and controls. Both the SNPs had no significant association with age of onset of illness, gender, disease location, disease behaviour, perianal disease, or extraintestinal manifestations of CD.
CONCLUSION Unlike observation form the West, polymorphisms in the PTPN2 gene (rs7234029 and rs2542151) are not associated with an increased risk of developing CD in Indian patients.
Collapse
Affiliation(s)
- Kaushik Chatterjee
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| | - Amit Kumar Dutta
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| | - Ashish Goel
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| | - Rekha Aaron
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| | - Vijayalekshmi Balakrishnan
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| | - Ajith Thomas
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| | - Anoop John
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| | - Rajeeb Jaleel
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| | - Deepu David
- Department of Gastroenterology, Christian Medical College, Vellore 632004, Tamil Nadu, India
| | - Reuben Thomas Kurien
- Department of Gastroenterology, Christian Medical College, Vellore 632004, Tamil Nadu, India
| | - SD Chowdhury
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| | - Ebby George Simon
- Department of Gastroenterology, Christian Medical College, Vellore 632004, Tamil Nadu, India
| | - AJ Joseph
- Department of Gastroenterology, Christian Medical College, Vellore 632004, Tamil Nadu, India
| | - Prasanna Premkumar
- Departments of Biostatistics, Christian Medical College, Vellore 632004, Tamil Nadu, India
| | - Anna B Pulimood
- Department of Gastrointestinal Sciences, Christian Medical College and Hospital, Vellore 632004, Tamil Nadu, India
| |
Collapse
|
17
|
Millrine D, Jenkins RH, Hughes STO, Jones SA. Making sense of IL-6 signalling cues in pathophysiology. FEBS Lett 2022; 596:567-588. [PMID: 34618359 PMCID: PMC9673051 DOI: 10.1002/1873-3468.14201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022]
Abstract
Unravelling the molecular mechanisms that account for functional pleiotropy is a major challenge for researchers in cytokine biology. Cytokine-receptor cross-reactivity and shared signalling pathways are considered primary drivers of cytokine pleiotropy. However, reports epitomized by studies of Jak-STAT cytokine signalling identify interesting biochemical and epigenetic determinants of transcription factor regulation that affect the delivery of signal-dependent cytokine responses. Here, a regulatory interplay between STAT transcription factors and their convergence to specific genomic enhancers support the fine-tuning of cytokine responses controlling host immunity, functional identity, and tissue homeostasis and repair. In this review, we provide an overview of the signalling networks that shape the way cells sense and interpret cytokine cues. With an emphasis on the biology of interleukin-6, we highlight the importance of these mechanisms to both physiological processes and pathophysiological outcomes.
Collapse
Affiliation(s)
- David Millrine
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
- Present address:
Medical Research Council Protein Phosphorylation and Ubiquitylation UnitSir James Black CentreSchool of Life SciencesUniversity of Dundee3rd FloorDundeeUK
| | - Robert H. Jenkins
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| | - Stuart T. O. Hughes
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| | - Simon A. Jones
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| |
Collapse
|
18
|
Goh PK, Wiede F, Zeissig MN, Britt KL, Liang S, Molloy T, Goode N, Xu R, Loi S, Muller M, Humbert PO, McLean C, Tiganis T. PTPN2 elicits cell autonomous and non-cell autonomous effects on antitumor immunity in triple-negative breast cancer. SCIENCE ADVANCES 2022; 8:eabk3338. [PMID: 35196085 PMCID: PMC8865802 DOI: 10.1126/sciadv.abk3338] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/24/2021] [Indexed: 05/22/2023]
Abstract
The tumor-suppressor PTPN2 is diminished in a subset of triple-negative breast cancers (TNBCs). Paradoxically, PTPN2-deficiency in tumors or T cells in mice can facilitate T cell recruitment and/or activation to promote antitumor immunity. Here, we explored the therapeutic potential of targeting PTPN2 in tumor cells and T cells. PTPN2-deficiency in TNBC associated with T cell infiltrates and PD-L1 expression, whereas low PTPN2 associated with improved survival. PTPN2 deletion in murine mammary epithelial cells TNBC models, did not promote tumorigenicity but increased STAT-1-dependent T cell recruitment and PD-L1 expression to repress tumor growth and enhance the efficacy of anti-PD-1. Furthermore, the combined deletion of PTPN2 in tumors and T cells facilitated T cell recruitment and activation and further repressed tumor growth or ablated tumors already predominated by exhausted T cells. Thus, PTPN2-targeting in tumors and/or T cells facilitates T cell recruitment and/or alleviates inhibitory constraints on T cells to combat TNBC.
Collapse
Affiliation(s)
- Pei Kee Goh
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Florian Wiede
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Mara N. Zeissig
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Kara L. Britt
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Shuwei Liang
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Tim Molloy
- St. Vincent’s Centre for Applied Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Nathan Goode
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Rachel Xu
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
| | - Mathias Muller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Patrick O. Humbert
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, 3010, Australia
- Research Centre for Molecular Cancer Prevention, La Trobe University, Melbourne, Victoria 3086, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria 3010, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Catriona McLean
- Anatomical Pathology, Alfred Hospital, Prahran, Victoria 3004, Australia
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Corresponding author.
| |
Collapse
|
19
|
Getahun A. Role of inhibitory signaling in peripheral B cell tolerance*. Immunol Rev 2022; 307:27-42. [PMID: 35128676 PMCID: PMC8986582 DOI: 10.1111/imr.13070] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022]
Abstract
At least 20% of B cells in the periphery expresses an antigen receptor with a degree of self-reactivity. If activated, these autoreactive B cells pose a risk as they can contribute to the development of autoimmune diseases. To prevent their activation, both B cell-intrinsic and extrinsic tolerance mechanisms are in place in healthy individuals. In this review article, I will focus on B cell-intrinsic mechanisms that prevent the activation of autoreactive B cells in the periphery. I will discuss how inhibitory signaling circuits are established in autoreactive B cells, focusing on the Lyn-SHIP-1-SHP-1 axis, how they contribute to peripheral immune tolerance, and how disruptions of these circuits can contribute to the development of autoimmunity.
Collapse
Affiliation(s)
- Andrew Getahun
- Department of Immunology and Microbiology University of Colorado SOM Aurora Colorado USA
- Department of Immunology and Genomic Medicine National Jewish Health Denver Colorado USA
| |
Collapse
|
20
|
The Frequency of Intrathyroidal Follicular Helper T Cells Varies with the Progression of Graves’ Disease and Hashimoto’s Thyroiditis. J Immunol Res 2022; 2022:4075522. [PMID: 35224111 PMCID: PMC8872690 DOI: 10.1155/2022/4075522] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 01/18/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Autoimmune thyroid diseases (AITD), mainly Graves' disease (GD) and Hashimoto's thyroiditis (HT), are common organ-specific autoimmune diseases characterized by circulating antibodies and lymphocyte infiltration. Follicular helper T (Tfh) cell dysregulation is involved in the development of autoimmune pathologies. We aimed to explore the role of intrathyroidal and circulating Tfh cells in patients with GD and HT. Methods Ultrasound-guided thyroid fine-needle aspiration (FNA) was conducted in 35 patients with GD, 40 patients with HT, and 22 patients with nonautoimmune thyroid disease (nAITD). Peripheral blood samples were also obtained from 40 patients with GD, 40 patients with HT, and 40 healthy controls. The frequencies of intrathyroidal and circulating Tfh cells from FNA and peripheral blood samples were assessed by flow cytometry. Additionally, the correlations between the frequencies of the Tfh cells and the levels of autoantibodies and hormones or disease duration were analyzed. Results The frequency of intrathyroidal CD4+CXCR5+ICOShigh Tfh cells was higher in HT patients than in GD patients. Significant correlations were identified between the percentages of circulating and intrathyroidal Tfh cells and the serum concentrations of thyroid autoantibodies, especially thyroglobulin antibodies (TgAb), in AITD. Intrathyroidal CD4+CXCR5+ICOShigh Tfh cells were positively correlated with free triiodothyronine (FT3) in HT patients but negatively correlated with FT3 in GD patients. In addition, HT patients with a longer disease duration had an increased frequency of intrathyroidal CD4+CXCR5+ICOShigh and CD4+CXCR5+PD-1+ Tfh cells. In contrast, in the GD patients, a longer disease duration did not affect the frequency of intrathyroidal CD4+CXCR5+ICOShigh but was associated with a lower frequency of CD4+CXCR5+PD-1+ Tfh cells. Conclusions Our data suggest that intrathyroidal Tfh cells might play a role in the pathogenesis of AITD and they are potential immunobiomarkers for AITD.
Collapse
|
21
|
Targeting PTPN22 does not enhance the efficacy of CAR T cells in solid tumours. Mol Cell Biol 2022; 42:e0044921. [PMID: 35041491 DOI: 10.1128/mcb.00449-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adoptive cell therapy with chimeric antigen receptor (CAR) T cells has revolutionised the treatment of certain B cell malignancies, but has been in ineffective against solid tumours. Recent studies have highlighted the potential of targeting negative regulators of T cell signalling to enhance the efficacy and extend the utility of CAR T cells to solid tumours. Autoimmunity-linked protein tyrosine phosphatase N22 (PTPN22) has been proposed as a target for cancer immunotherapy. Here we have used CRISPR/Cas9 gene-editing to generate PTPN22-deficient (Ptpn22Δ/Δ) mice (C57BL/6) and assessed the impact of PTPN22 deficiency on the cytotoxicity and efficacy of CAR T cells in vitro and in vivo. As reported previously, PTPN22 deficiency was accompanied by the promotion of effector T cell responses ex vivo and the repression of syngeneic tumour growth in vivo. However, PTPN22-deficiency did not enhance the cytotoxic activity of murine CAR T cells targeting the extracellular domain of the human oncoprotein HER2 in vitro. Moreover, PTPN22-deficient α-HER2 CAR T cells or ovalbumin-specific OT-I CD8+ T cells adoptively transferred into mice bearing HER2+ mammary tumours or ovalbumin-expressing mammary or colorectal tumours respectively were no more effective than their wild type counterparts in suppressing tumour growth. The deletion of PTPN22 using CRISPR/Cas9 gene-editing also did not affect the cytotoxic activity of human CAR T cells targeting the Lewis Y antigen that is expressed by many human solid tumours. Therefore, PTPN22-deficiency does not enhance the anti-tumour activity of CAR T cells in solid organ malignancies.
Collapse
|
22
|
Wiede F, Lu KH, Du X, Zeissig MN, Xu R, Goh PK, Xirouchaki CE, Hogarth SJ, Greatorex S, Sek K, Daly RJ, Beavis PA, Darcy PK, Tonks NK, Tiganis T. PTP1B is an intracellular checkpoint that limits T cell and CAR T cell anti-tumor immunity. Cancer Discov 2021; 12:752-773. [PMID: 34794959 DOI: 10.1158/2159-8290.cd-21-0694] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/01/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022]
Abstract
Immunotherapies aimed at alleviating the inhibitory constraints on Tcells have revolutionised cancer management. To date, these have focused on the blockade of cell surface checkpoints such as PD-1. Herein we identify protein-tyrosine-phosphatase-1B (PTP1B) as an intracellular checkpoint that is upregulated in T cells in tumors. We show that the increased PTP1B limits T cell expansion and cytotoxicity to contribute to tumor growth. T cell-specific PTP1B deletion increased STAT-5 signaling and this enhanced the antigen-induced expansion and cytotoxicity of CD8+ T cells to suppress tumor growth. The pharmacological inhibition of PTP1B recapitulated the T cell-mediated repression of tumor growth and enhanced the response to PD-1 blockade. Furthermore, the deletion or inhibition of PTP1B enhanced the efficacy of adoptively-transferred chimeric-antigen-receptor (CAR) T cells against solid tumors. Our findings identify PTP1B as an intracellular checkpoint whose inhibition can alleviate the inhibitory constraints on T cells and CAR T cells to combat cancer.
Collapse
Affiliation(s)
- Florian Wiede
- Biochemistry and Molecular Biology, Monash University
| | - Kun-Hui Lu
- Cancer Research, Peter MacCallum Cancer Centre
| | - Xin Du
- Peter MacCallum Cancer Centre
| | | | | | - Pei Kee Goh
- Biochemistry and Molecular Biology, Monash University
| | | | | | | | - Kevin Sek
- Cancer Immunology Program, Peter MacCallum Cancer Research Centre
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Research Centre
| | | | | | - Tony Tiganis
- Biochemistry and Molecular Biology, Monash University
| |
Collapse
|
23
|
Zhao M, Jia S, Gao X, Qiu H, Wu R, Wu H, Lu Q. Comparative Analysis of Global Proteome and Lysine Acetylome Between Naive CD4 + T Cells and CD4 + T Follicular Helper Cells. Front Immunol 2021; 12:643441. [PMID: 33841426 PMCID: PMC8027069 DOI: 10.3389/fimmu.2021.643441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/19/2021] [Indexed: 12/05/2022] Open
Abstract
As a subgroup of CD4+ T helper cells, follicular helper T (Tfh) cells provide help to germinal center B cells and mediate the development of long-lived humoral immunity. Dysregulation of Tfh cells is associated with several major autoimmune diseases. Although recent studies showed that Tfh cell differentiation is controlled by the transcription factor Bcl6, cytokines, and cell-cell signals, limited information is available on the proteome and post-translational modifications (PTMs) of proteins in human Tfh cells. In the present study, we investigated quantitative proteome and acetylome in human naive CD4+ T cells and in vitro induced Tfh (iTfh) cells using the tandem mass tag (TMT) labeling technique, antibody-based affinity enrichment, and high-resolution liquid chromatography-mass spectrometry (LC-MS)/mass spectrometry (MS) analysis. In total, we identified 802 upregulated proteins and 598 downregulated proteins at the threshold of 1.5-fold in iTfh cells compared to naive CD4+ T cells. With the aid of intensive bioinformatics, the biological process, the cellular compartment, the molecular function, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and protein–protein interaction of these differentially expressed proteins were revealed. Moreover, the acetylome data showed that 22 lysine (K) acetylated proteins are upregulated and 26 K acetylated proteins are downregulated in iTfh cells compared to the naive CD4+ T cells, among which 11 differentially acetylated K residues in core histones were identified, indicating that protein acetylation and epigenetic mechanism are involved in regulating Tfh cell differentiation. The study provides some important clues for investigating T cell activation and Tfh cell differentiation.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Key Technologies of Diagnosis and Treatment for Immune-Related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Sujie Jia
- Department of Pharmaceutics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiaofei Gao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Key Technologies of Diagnosis and Treatment for Immune-Related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Hong Qiu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Key Technologies of Diagnosis and Treatment for Immune-Related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Ruifang Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Key Technologies of Diagnosis and Treatment for Immune-Related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Unit of Key Technologies of Diagnosis and Treatment for Immune-Related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
24
|
Wang YN, Liu S, Jia T, Feng Y, Zhang W, Xu X, Zhang D. T Cell Protein Tyrosine Phosphatase in Osteoimmunology. Front Immunol 2021; 12:620333. [PMID: 33692794 PMCID: PMC7938726 DOI: 10.3389/fimmu.2021.620333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/04/2021] [Indexed: 12/23/2022] Open
Abstract
Osteoimmunology highlights the two-way communication between bone and immune cells. T cell protein tyrosine phosphatase (TCPTP), also known as protein-tyrosine phosphatase non-receptor 2 (PTPN2), is an intracellular protein tyrosine phosphatase (PTP) essential in regulating immune responses and bone metabolism via dephosphorylating target proteins. Tcptp knockout in systemic or specific immune cells can seriously damage the immune function, resulting in bone metabolism disorders. This review provided fresh insights into the potential role of TCPTP in osteoimmunology. Overall, the regulation of osteoimmunology by TCPTP is extremely complicated. TCPTP negatively regulates macrophages activation and inflammatory factors secretion to inhibit bone resorption. TCPTP regulates T lymphocytes differentiation and T lymphocytes-related cytokines signaling to maintain bone homeostasis. TCPTP is also expected to regulate bone metabolism by targeting B lymphocytes under certain time and conditions. This review offers a comprehensive update on the roles of TCPTP in osteoimmunology, which can be a promising target for the prevention and treatment of inflammatory bone loss.
Collapse
Affiliation(s)
- Ya-Nan Wang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Shiyue Liu
- Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China.,Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tingting Jia
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Yao Feng
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Wenjing Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Dongjiao Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
25
|
Huang Z, Liu M, Li D, Tan Y, Zhang R, Xia Z, Wang P, Jiao B, Liu P, Ren R. PTPN2 regulates the activation of KRAS and plays a critical role in proliferation and survival of KRAS-driven cancer cells. J Biol Chem 2020; 295:18343-18354. [PMID: 33122197 PMCID: PMC7939389 DOI: 10.1074/jbc.ra119.011060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
RAS genes are the most commonly mutated in human cancers and play critical roles in tumor initiation, progression, and drug resistance. Identification of targets that block RAS signaling is pivotal to develop therapies for RAS-related cancer. As RAS translocation to the plasma membrane (PM) is essential for its effective signal transduction, we devised a high-content screening assay to search for genes regulating KRAS membrane association. We found that the tyrosine phosphatase PTPN2 regulates the plasma membrane localization of KRAS. Knockdown of PTPN2 reduced the proliferation and promoted apoptosis in KRAS-dependent cancer cells, but not in KRAS-independent cells. Mechanistically, PTPN2 negatively regulates tyrosine phosphorylation of KRAS, which, in turn, affects the activation KRAS and its downstream signaling. Consistently, analysis of the TCGA database demonstrates that high expression of PTPN2 is significantly associated with poor prognosis of patients with KRAS-mutant pancreatic adenocarcinoma. These results indicate that PTPN2 is a key regulator of KRAS and may serve as a new target for therapy of KRAS-driven cancer.
Collapse
Affiliation(s)
- Zhangsen Huang
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingzhu Liu
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Donghe Li
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Tan
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruihong Zhang
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhizhou Xia
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peihong Wang
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Jiao
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Liu
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ruibao Ren
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine at Shanghai, Collaborative Innovation Center of Hematology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
26
|
Castro-Sanchez P, Teagle AR, Prade S, Zamoyska R. Modulation of TCR Signaling by Tyrosine Phosphatases: From Autoimmunity to Immunotherapy. Front Cell Dev Biol 2020; 8:608747. [PMID: 33425916 PMCID: PMC7793860 DOI: 10.3389/fcell.2020.608747] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Early TCR signaling is dependent on rapid phosphorylation and dephosphorylation of multiple signaling and adaptor proteins, leading to T cell activation. This process is tightly regulated by an intricate web of interactions between kinases and phosphatases. A number of tyrosine phosphatases have been shown to modulate T cell responses and thus alter T cell fate by negatively regulating early TCR signaling. Mutations in some of these enzymes are associated with enhanced predisposition to autoimmunity in humans, and mouse models deficient in orthologous genes often show T cell hyper-activation. Therefore, phosphatases are emerging as potential targets in situations where it is desirable to enhance T cell responses, such as immune responses to tumors. In this review, we summarize the current knowledge about tyrosine phosphatases that regulate early TCR signaling and discuss their involvement in autoimmunity and their potential as targets for tumor immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Rose Zamoyska
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
27
|
Dodd GT, Xirouchaki CE, Eramo M, Mitchell CA, Andrews ZB, Henry BA, Cowley MA, Tiganis T. Intranasal Targeting of Hypothalamic PTP1B and TCPTP Reinstates Leptin and Insulin Sensitivity and Promotes Weight Loss in Obesity. Cell Rep 2020; 28:2905-2922.e5. [PMID: 31509751 DOI: 10.1016/j.celrep.2019.08.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/29/2019] [Accepted: 08/02/2019] [Indexed: 12/11/2022] Open
Abstract
The importance of hypothalamic leptin and insulin resistance in the development and maintenance of obesity remains unclear. The tyrosine phosphatases protein tyrosine phosphatase 1B (PTP1B) and T cell protein tyrosine phosphatase (TCPTP) attenuate leptin and insulin signaling and are elevated in the hypothalami of obese mice. We report that elevated PTP1B and TCPTP antagonize hypothalamic leptin and insulin signaling and contribute to the maintenance of obesity. Deletion of PTP1B and TCPTP in the hypothalami of obese mice enhances CNS leptin and insulin sensitivity, represses feeding, and increases browning, to decrease adiposity and improve glucose metabolism. The daily intranasal administration of a PTP1B inhibitor, plus the glucocorticoid antagonist RU486 that decreases TCPTP expression, represses feeding, increases browning, promotes weight loss, and improves glucose metabolism in obese mice. Our findings causally link heightened hypothalamic PTP1B and TCPTP with leptin and insulin resistance and the maintenance of obesity and define a viable pharmacological approach by which to promote weight loss in obesity.
Collapse
Affiliation(s)
- Garron T Dodd
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Chrysovalantou E Xirouchaki
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Matthew Eramo
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Christina A Mitchell
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Zane B Andrews
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Physiology, Monash University, VIC 3800, Australia
| | - Belinda A Henry
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Physiology, Monash University, VIC 3800, Australia
| | - Michael A Cowley
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Physiology, Monash University, VIC 3800, Australia
| | - Tony Tiganis
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Monash Metabolic Phenotyping Facility, Monash University, VIC, Australia; Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.
| |
Collapse
|
28
|
Cordes F, Foell D, Ding JN, Varga G, Bettenworth D. Differential regulation of JAK/STAT-signaling in patients with ulcerative colitis and Crohn's disease. World J Gastroenterol 2020; 26:4055-4075. [PMID: 32821070 PMCID: PMC7403801 DOI: 10.3748/wjg.v26.i28.4055] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/24/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
In 2018, the pan-Janus kinase (JAK) inhibitor tofacitinib was launched for the treatment of ulcerative colitis (UC). Although tofacitinib has proven efficacious in patients with active UC, it failed in patients with Crohn's disease (CD). This finding strongly hints at a different contribution of JAK signaling in both entities. Here, we review the current knowledge on the interplay between the JAK/signal transducer and activator of transcription (STAT) pathway and inflammatory bowel diseases (IBD). In particular, we provide a detailed overview of the differences and similarities of JAK/STAT-signaling in UC and CD, highlight the impact of the JAK/STAT pathway in experimental colitis models and summarize the published evidence on JAK/STAT-signaling in immune cells of IBD as well as the genetic association between the JAK/STAT pathway and IBD. Finally, we describe novel treatment strategies targeting JAK/STAT inhibition in UC and CD and comment on the limitations and challenges of the new drug class.
Collapse
Affiliation(s)
- Friederike Cordes
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster D-48149, Germany
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Münster, Münster D-48149, Germany
| | - John Nik Ding
- Department of Gastroenterology, St. Vincent’s Hospital, Melbourne 3002, Australia
- Department of Medicine, University of Melbourne, East Melbourne 3002, Australia
| | - Georg Varga
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Münster, Münster D-48149, Germany
| | - Dominik Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster D-48149, Germany
| |
Collapse
|
29
|
Thaventhiran JED, Lango Allen H, Burren OS, Rae W, Greene D, Staples E, Zhang Z, Farmery JHR, Simeoni I, Rivers E, Maimaris J, Penkett CJ, Stephens J, Deevi SVV, Sanchis-Juan A, Gleadall NS, Thomas MJ, Sargur RB, Gordins P, Baxendale HE, Brown M, Tuijnenburg P, Worth A, Hanson S, Linger RJ, Buckland MS, Rayner-Matthews PJ, Gilmour KC, Samarghitean C, Seneviratne SL, Sansom DM, Lynch AG, Megy K, Ellinghaus E, Ellinghaus D, Jorgensen SF, Karlsen TH, Stirrups KE, Cutler AJ, Kumararatne DS, Chandra A, Edgar JDM, Herwadkar A, Cooper N, Grigoriadou S, Huissoon AP, Goddard S, Jolles S, Schuetz C, Boschann F, Lyons PA, Hurles ME, Savic S, Burns SO, Kuijpers TW, Turro E, Ouwehand WH, Thrasher AJ, Smith KGC. Whole-genome sequencing of a sporadic primary immunodeficiency cohort. Nature 2020; 583:90-95. [PMID: 32499645 PMCID: PMC7334047 DOI: 10.1038/s41586-020-2265-1] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 02/26/2020] [Indexed: 12/19/2022]
Abstract
Primary immunodeficiency (PID) is characterized by recurrent and often life-threatening infections, autoimmunity and cancer, and it poses major diagnostic and therapeutic challenges. Although the most severe forms of PID are identified in early childhood, most patients present in adulthood, typically with no apparent family history and a variable clinical phenotype of widespread immune dysregulation: about 25% of patients have autoimmune disease, allergy is prevalent and up to 10% develop lymphoid malignancies1-3. Consequently, in sporadic (or non-familial) PID genetic diagnosis is difficult and the role of genetics is not well defined. Here we address these challenges by performing whole-genome sequencing in a large PID cohort of 1,318 participants. An analysis of the coding regions of the genome in 886 index cases of PID found that disease-causing mutations in known genes that are implicated in monogenic PID occurred in 10.3% of these patients, and a Bayesian approach (BeviMed4) identified multiple new candidate PID-associated genes, including IVNS1ABP. We also examined the noncoding genome, and found deletions in regulatory regions that contribute to disease causation. In addition, we used a genome-wide association study to identify loci that are associated with PID, and found evidence for the colocalization of-and interplay between-novel high-penetrance monogenic variants and common variants (at the PTPN2 and SOCS1 loci). This begins to explain the contribution of common variants to the variable penetrance and phenotypic complexity that are observed in PID. Thus, using a cohort-based whole-genome-sequencing approach in the diagnosis of PID can increase diagnostic yield and further our understanding of the key pathways that influence immune responsiveness in humans.
Collapse
Affiliation(s)
- James E D Thaventhiran
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK.
- Medical Research Council Toxicology Unit, School of Biological Sciences, University of Cambridge, Cambridge, UK.
| | - Hana Lango Allen
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Oliver S Burren
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - William Rae
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Daniel Greene
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge, UK
| | - Emily Staples
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Zinan Zhang
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology and Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - James H R Farmery
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Ilenia Simeoni
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Elizabeth Rivers
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Jesmeen Maimaris
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Christopher J Penkett
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Jonathan Stephens
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Sri V V Deevi
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Alba Sanchis-Juan
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicholas S Gleadall
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Moira J Thomas
- Department of Immunology, Queen Elizabeth University Hospital, Glasgow, UK
- Gartnavel General Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Ravishankar B Sargur
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Pavels Gordins
- East Yorkshire Regional Adult Immunology and Allergy Unit, Hull Royal Infirmary, Hull and East Yorkshire Hospitals NHS Trust, Hull, UK
| | - Helen E Baxendale
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Matthew Brown
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Paul Tuijnenburg
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Austen Worth
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Steven Hanson
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Rachel J Linger
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Matthew S Buckland
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Paula J Rayner-Matthews
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Kimberly C Gilmour
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Crina Samarghitean
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Suranjith L Seneviratne
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - David M Sansom
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Andy G Lynch
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
- School of Mathematics and Statistics, University of St Andrews, St Andrews, UK
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Karyn Megy
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Eva Ellinghaus
- K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital, Oslo, Norway
| | - David Ellinghaus
- Department of Transplantation, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Silje F Jorgensen
- Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Oslo, Norway
| | - Tom H Karlsen
- K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo University Hospital, Oslo, Norway
| | - Kathleen E Stirrups
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Antony J Cutler
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Dinakantha S Kumararatne
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - Anita Chandra
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
| | - J David M Edgar
- St James's Hospital, Dublin, Ireland
- Trinity College Dublin, Dublin, Ireland
| | | | - Nichola Cooper
- Department of Medicine, Imperial College London, London, UK
| | | | - Aarnoud P Huissoon
- West Midlands Immunodeficiency Centre, University Hospitals Birmingham, Birmingham, UK
- Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Sarah Goddard
- University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - Catharina Schuetz
- Department of Pediatric Immunology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Felix Boschann
- Institute of Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthew E Hurles
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Sinisa Savic
- Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, UK
- The NIHR Leeds Biomedical Research Centre, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
| | - Siobhan O Burns
- Institute of Immunity and Transplantation, University College London, London, UK
- Department of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- Department of Blood Cell Research, Sanquin, Amsterdam, The Netherlands
| | - Ernest Turro
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge, UK
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Adrian J Thrasher
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
30
|
Chen J, Zhao X, Yuan Y, Jing JJ. The expression patterns and the diagnostic/prognostic roles of PTPN family members in digestive tract cancers. Cancer Cell Int 2020; 20:238. [PMID: 32536826 PMCID: PMC7291430 DOI: 10.1186/s12935-020-01315-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background Non-receptor protein tyrosine phosphatases (PTPNs) are a set of enzymes involved in the tyrosyl phosphorylation. The present study intended to clarify the associations between the expression patterns of PTPN family members, and diagnosis as well as the prognosis of digestive tract cancers. Methods Oncomine and Ualcan were used to analyze PTPN expressions. Data from The Cancer Genome Atlas (TCGA) were downloaded through UCSC Xena for validation and to explore the relationship of the PTPN expression with diagnosis, clinicopathological parameters and survival of digestive tract cancers. Gene ontology enrichment analysis was conducted using the DAVID database. The gene–gene interaction network was performed by GeneMANIA and the protein–protein interaction (PPI) network was built using STRING portal coupled with Cytoscape. The expression of differentially expressed PTPNs in cancer cell lines were explored using CCLE. Moreover, by histological verification, the expression of four PTPNs in digestive tract cancers were further analyzed. Results Most PTPN family members were associated with digestive tract cancers according to Oncomine, Ualcan and TCGA data. Several PTPN members were differentially expressed in digestive tract cancers. For esophageal carcinoma (ESCA), PTPN1 and PTPN12 levels were correlated with incidence; PTPN20 was associated with poor prognosis. For stomach adenocarcinoma (STAD), PTPN2 and PTPN12 levels were correlated with incidence; PTPN3, PTPN5, PTPN7, PTPN11, PTPN13, PTPN14, PTPN18 and PTPN23 were correlated with pathological grade; PTPN20 expression was related with both TNM stage and N stage; PTPN22 was associated with T stage and pathological grade; decreased expression of PTPN5 and PTPN13 implied worse overall survival of STAD, while elevated PTPN6 expression indicated better prognosis. For colorectal cancer (CRC), PTPN2, PTPN21 and PTPN22 levels were correlated with incidence; expression of PTPN5, PTPN12, and PTPN14 was correlated with TNM stage and N stage; high PTPN5 or PTPN7 expression was associated with increased hazards of death. CCLE analyses showed that in esophagus cancer cell lines, PTPN1, PTPN4 and PTPN12 were highly expressed; in gastric cancer cell lines, PTPN2 and PTPN12 were highly expressed; in colorectal cancer cell lines, PTPN12 was highly expressed while PTPN22 was downregulated. Results of histological verification experiment showed differential expressions of PTPN22 in CRC, and PTPN12 in GC and CRC. Conclusions Members of PTPN family were differentially expressed in digestive tract cancers. Correlations were found between PTPN genes and clinicopathological parameters of patients. Expression of PTPN12 was upregulated in both STAD and CRC, and thus could be used as a diagnostic biomarker. Differential expression of PTPN12 in GC and CRC, and PTPN22 in CRC were presented in our histological verification experiment.
Collapse
Affiliation(s)
- Jing Chen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning People's Republic of China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, 110001 China.,Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001 China
| | - Xu Zhao
- Mathematical Computer Teaching and Research Office, Liaoning Vocational College of Medicine, Shenyang, 110101 China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning People's Republic of China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, 110001 China.,Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001 China
| | - Jing-Jing Jing
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning People's Republic of China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, 110001 China.,Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001 China
| |
Collapse
|
31
|
Zhang T, He L, Wang Z, Dong W, Sun W, Qin Y, Zhang P, Zhang H. Calcitriol enhances Doxorubicin-induced apoptosis in papillary thyroid carcinoma cells via regulating VDR/PTPN2/p-STAT3 pathway. J Cell Mol Med 2020; 24:5629-5639. [PMID: 32285621 PMCID: PMC7214146 DOI: 10.1111/jcmm.15224] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 02/28/2020] [Accepted: 03/13/2020] [Indexed: 12/29/2022] Open
Abstract
There is increasing evidence that vitamin D deficiency is the risk factor for multiple diseases, such as immune disorder, cardiovascular disease and cancer. Calcitriol is the active form of vitamin D with beneficial effects on anti‐cancer by binding vitamin D receptor (VDR). The primary aim of this study was to investigate the role of Calcitriol on papillary thyroid carcinoma (PTC) and explore the possible mechanism. We found nuclear VDR expression in PTC samples was negatively correlated with STAT3 hyperphosphorylation that indicated worse PTC clinicopathologic characteristics. Calcitriol treatment up‐regulated VDR and protein tyrosine phosphatase N 2 (PTPN2) expression, down‐regulated signal transducers and activators of transcription (STAT3) phosphorylation and thereby facilitating chemotherapy drug Doxorubicin‐induced apoptosis in PTC cell lines. However, the apoptosis‐promoting effect of Calcitriol and Doxorubicin co‐treatment was abrogated by STAT3 hyperphosphorylation, indicating suppression of STAT3 phosphorylation was essential for combined treatment of Calcitriol and Doxorubicin in PTC. Together, these results suggested that Calcitriol reinforced the sensitivity of PTC cells to Doxorubicin by regulating VDR/PTPN2/p‐STAT3 signalling pathway.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Liang He
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Zhihong Wang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wenwu Dong
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wei Sun
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yuan Qin
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Ping Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
32
|
Wiede F, Lu K, Du X, Liang S, Hochheiser K, Dodd GT, Goh PK, Kearney C, Meyran D, Beavis PA, Henderson MA, Park SL, Waithman J, Zhang S, Zhang Z, Oliaro J, Gebhardt T, Darcy PK, Tiganis T. PTPN2 phosphatase deletion in T cells promotes anti-tumour immunity and CAR T-cell efficacy in solid tumours. EMBO J 2020; 39:e103637. [PMID: 31803974 PMCID: PMC6960448 DOI: 10.15252/embj.2019103637] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/04/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022] Open
Abstract
Although adoptive T-cell therapy has shown remarkable clinical efficacy in haematological malignancies, its success in combating solid tumours has been limited. Here, we report that PTPN2 deletion in T cells enhances cancer immunosurveillance and the efficacy of adoptively transferred tumour-specific T cells. T-cell-specific PTPN2 deficiency prevented tumours forming in aged mice heterozygous for the tumour suppressor p53. Adoptive transfer of PTPN2-deficient CD8+ T cells markedly repressed tumour formation in mice bearing mammary tumours. Moreover, PTPN2 deletion in T cells expressing a chimeric antigen receptor (CAR) specific for the oncoprotein HER-2 increased the activation of the Src family kinase LCK and cytokine-induced STAT-5 signalling, thereby enhancing both CAR T-cell activation and homing to CXCL9/10-expressing tumours to eradicate HER-2+ mammary tumours in vivo. Our findings define PTPN2 as a target for bolstering T-cell-mediated anti-tumour immunity and CAR T-cell therapy against solid tumours.
Collapse
Affiliation(s)
- Florian Wiede
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | - Kun‐Hui Lu
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | - Xin Du
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | - Shuwei Liang
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | - Katharina Hochheiser
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Department of Microbiology and ImmunologyThe University of MelbourneMelbourneVic.Australia
- Peter Doherty Institute for Infection and ImmunityMelbourneVic.Australia
| | - Garron T Dodd
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
| | - Pei K Goh
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | | | | | - Paul A Beavis
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | | | - Simone L Park
- Department of Microbiology and ImmunologyThe University of MelbourneMelbourneVic.Australia
- Peter Doherty Institute for Infection and ImmunityMelbourneVic.Australia
| | - Jason Waithman
- Telethon Kids InstituteUniversity of Western AustraliaPerthWAAustralia
| | - Sheng Zhang
- Department of Medicinal Chemistry and Molecular PharmacologyInstitute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Zhong‐Yin Zhang
- Department of Medicinal Chemistry and Molecular PharmacologyInstitute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Jane Oliaro
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | - Thomas Gebhardt
- Department of Microbiology and ImmunologyThe University of MelbourneMelbourneVic.Australia
- Peter Doherty Institute for Infection and ImmunityMelbourneVic.Australia
| | - Phillip K Darcy
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | - Tony Tiganis
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| |
Collapse
|
33
|
Shao F, Zheng P, Yu D, Zhou Z, Jia L. Follicular helper T cells in type 1 diabetes. FASEB J 2019; 34:30-40. [PMID: 31914661 DOI: 10.1096/fj.201901637r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/09/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Feng Shao
- Department of Metabolism & Endocrinology The Second Xiangya HospitalCentral South University Changsha China
- Key Laboratory of Diabetes Immunology Central South University, Ministry of Education, National Clinical Research Center for Metabolic Diseases Changsha China
| | - Peilin Zheng
- Department of Endocrinology, Shenzhen People’s Hospital The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology Shenzhen China
| | - Di Yu
- The University of Queensland Diamantina Institute, Translational Research Institute Brisbane Queensland Australia
- Shandong Analysis and Test Center Shandong Academy of Sciences Jinan China
- China‐Australia Centre for Personalised Immunology Shanghai Renji Hospital Shanghai Jiaotong University School of Medicine Shanghai China
| | - Zhiguang Zhou
- Department of Metabolism & Endocrinology The Second Xiangya HospitalCentral South University Changsha China
- Key Laboratory of Diabetes Immunology Central South University, Ministry of Education, National Clinical Research Center for Metabolic Diseases Changsha China
| | - Lijing Jia
- Department of Endocrinology, Shenzhen People’s Hospital The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology Shenzhen China
| |
Collapse
|
34
|
Ilonen J, Lempainen J, Veijola R. The heterogeneous pathogenesis of type 1 diabetes mellitus. Nat Rev Endocrinol 2019; 15:635-650. [PMID: 31534209 DOI: 10.1038/s41574-019-0254-y] [Citation(s) in RCA: 279] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/13/2019] [Indexed: 12/14/2022]
Abstract
Type 1 diabetes mellitus (T1DM) results from the destruction of pancreatic β-cells that is mediated by the immune system. Multiple genetic and environmental factors found in variable combinations in individual patients are involved in the development of T1DM. Genetic risk is defined by the presence of particular allele combinations, which in the major susceptibility locus (the HLA region) affect T cell recognition and tolerance to foreign and autologous molecules. Multiple other loci also regulate and affect features of specific immune responses and modify the vulnerability of β-cells to inflammatory mediators. Compared with the genetic factors, environmental factors that affect the development of T1DM are less well characterized but contact with particular microorganisms is emerging as an important factor. Certain infections might affect immune regulation, and the role of commensal microorganisms, such as the gut microbiota, are important in the education of the developing immune system. Some evidence also suggests that nutritional factors are important. Multiple islet-specific autoantibodies are found in the circulation from a few weeks to up to 20 years before the onset of clinical disease and this prediabetic phase provides a potential opportunity to manipulate the islet-specific immune response to prevent or postpone β-cell loss. The latest developments in understanding the heterogeneity of T1DM and characterization of major disease subtypes might help in the development of preventive treatments.
Collapse
Affiliation(s)
- Jorma Ilonen
- Institue of Biomedicine, University of Turku and Clinical Microbiology, Turku University Hospital, Turku, Finland.
| | - Johanna Lempainen
- Institue of Biomedicine, University of Turku and Clinical Microbiology, Turku University Hospital, Turku, Finland
- Department of Paediatrics, University of Turku and Turku University Hospital, Turku, Finland
| | - Riitta Veijola
- Department of Paediatrics, University of Oulu and Oulu University Hospital, Oulu, Finland
| |
Collapse
|
35
|
Castro-Sánchez P, Aguilar-Sopeña O, Alegre-Gómez S, Ramirez-Munoz R, Roda-Navarro P. Regulation of CD4 + T Cell Signaling and Immunological Synapse by Protein Tyrosine Phosphatases: Molecular Mechanisms in Autoimmunity. Front Immunol 2019; 10:1447. [PMID: 31297117 PMCID: PMC6607956 DOI: 10.3389/fimmu.2019.01447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
T cell activation and effector function is mediated by the formation of a long-lasting interaction established between T cells and antigen-presenting cells (APCs) called immunological synapse (IS). During T cell activation, different signaling molecules as well as the cytoskeleton and the endosomal compartment are polarized to the IS. This molecular dynamics is tightly regulated by phosphorylation networks, which are controlled by protein tyrosine phosphatases (PTPs). While some PTPs are known to be important regulators of adhesion, ligand discrimination or the stimulation threshold, there is still little information about the regulatory role of PTPs in cytoskeleton rearrangements and endosomal compartment dynamics. Besides, spatial and temporal regulation of PTPs and substrates at the IS is only barely known. Consistent with an important role of PTPs in T cell activation, multiple mutations as well as altered expression levels or dynamic behaviors have been associated with autoimmune diseases. However, the precise mechanism for the regulation of T cell activation and effector function by PTPs in health and autoimmunity is not fully understood. Herein, we review the current knowledge about the regulatory role of PTPs in CD4+ T cell activation, IS assembly and effector function. The potential molecular mechanisms mediating the action of these enzymes in autoimmune disorders are discussed.
Collapse
Affiliation(s)
- Patricia Castro-Sánchez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| | - Oscar Aguilar-Sopeña
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| | - Sergio Alegre-Gómez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| | - Rocio Ramirez-Munoz
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| | - Pedro Roda-Navarro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| |
Collapse
|
36
|
Wiede F, Brodnicki TC, Goh PK, Leong YA, Jones GW, Yu D, Baxter AG, Jones SA, Kay TWH, Tiganis T. T-Cell-Specific PTPN2 Deficiency in NOD Mice Accelerates the Development of Type 1 Diabetes and Autoimmune Comorbidities. Diabetes 2019; 68:1251-1266. [PMID: 30936146 DOI: 10.2337/db18-1362] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 03/17/2019] [Indexed: 11/13/2022]
Abstract
Genome-wide association studies have identified PTPN2 as an important non-MHC gene for autoimmunity. Single nucleotide polymorphisms that reduce PTPN2 expression have been linked with the development of various autoimmune disorders, including type 1 diabetes. The tyrosine phosphatase PTPN2 attenuates T-cell receptor and cytokine signaling in T cells to maintain peripheral tolerance, but the extent to which PTPN2 deficiency in T cells might influence type 1 diabetes onset remains unclear. NOD mice develop spontaneous autoimmune type 1 diabetes similar to that seen in humans. In this study, T-cell PTPN2 deficiency in NOD mice markedly accelerated the onset and increased the incidence of type 1 diabetes as well as that of other disorders, including colitis and Sjögren syndrome. Although PTPN2 deficiency in CD8+ T cells alone was able to drive the destruction of pancreatic β-cells and the onset of diabetes, T-cell-specific PTPN2 deficiency was also accompanied by increased CD4+ T-helper type 1 differentiation and T-follicular-helper cell polarization and increased the abundance of B cells in pancreatic islets as seen in human type 1 diabetes. These findings causally link PTPN2 deficiency in T cells with the development of type 1 diabetes and associated autoimmune comorbidities.
Collapse
Affiliation(s)
- Florian Wiede
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Thomas C Brodnicki
- St. Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| | - Pei Kee Goh
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Yew A Leong
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Gareth W Jones
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
- Systems Immunity University Research Institute, Cardiff University, Cardiff, U.K
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, U.K
| | - Di Yu
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Alan G Baxter
- Comparative Genomics Centre, James Cook University, Townsville, Queensland, Australia
| | - Simon A Jones
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
- Systems Immunity University Research Institute, Cardiff University, Cardiff, U.K
| | - Thomas W H Kay
- St. Vincent's Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Chen S, Fan H, Feng Y, Zhang Y, Chen Y, Gu Y, Shi Y, Dai H, Zhang M, Xu X, Chen H, Yang T, Xu K. The association between rs1893217, rs478582 in PTPN2 and T1D risk with different diagnosed age, and related clinical characteristics in Chinese Han population. Autoimmunity 2019; 52:95-101. [PMID: 31030572 DOI: 10.1080/08916934.2019.1608191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To investigate the association between polymorphisms in PTPN2 (rs1893217 and rs478582) and type 1 diabetes (T1D) risk with different diagnosed age, as well as related clinical characteristics in Chinese Han population. METHODS A total of 2270 Chinese Han individuals (1023 T1D patients and 1247 healthy controls) were genotyped for rs1893217 and rs478582. And 306 newly diagnosed T1D patients were measured for C-peptide levels based on a standard mixed-meal tolerance test. In addition, 40 healthy controls were analyzed for different T cell subsets by multi-color flow cytometry. RESULTS Neither rs1893217 nor rs478582 showed any association with T1D risk under an additive model. Stratified analysis for T1D diagnosed age revealed that rs1893217, but not rs478582, was significantly associated with T1D patients diagnosed age ≤18 (OR =0.80, 95% CI: 0.67-0.97, p = 0.02). For those diagnosed age >18, neither of them showed any association. We also found that rs1893217 had a higher positive rate of ZnT8A (CC vs. TT carrier, OR = 2.07, 95% CI: 1.07-4.03, p = 0.026) and IA-2A (CT vs. TT carrier, OR = 1.36, 95% CI: 1.02-1.80, p = 0.038). Furthermore, for rs478582, compared with TT, healthy individuals carrying CC/CT carriers had significantly lower frequency and Helios expression of naive Treg subsets (p = 0.049 and 0.048 respectively), but not secreting or activating Treg subsets. In addition, we did not find any association between these two polymorphisms and residual β-cell function in newly diagnosed T1D patients. CONCLUSIONS Our results suggest that rs1893217 may increase the risk of early-onset T1D and affect humoral immunity, while rs478582 may affect Treg subsets.
Collapse
Affiliation(s)
- Shu Chen
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Hongqi Fan
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Yingjie Feng
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Yuyue Zhang
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Yang Chen
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Yong Gu
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Yun Shi
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Hao Dai
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Mei Zhang
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Xinyu Xu
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Heng Chen
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Tao Yang
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Kuanfeng Xu
- a Department of Endocrinology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| |
Collapse
|
38
|
LaFleur MW, Nguyen TH, Coxe MA, Yates KB, Trombley JD, Weiss SA, Brown FD, Gillis JE, Coxe DJ, Doench JG, Haining WN, Sharpe AH. A CRISPR-Cas9 delivery system for in vivo screening of genes in the immune system. Nat Commun 2019; 10:1668. [PMID: 30971695 PMCID: PMC6458184 DOI: 10.1038/s41467-019-09656-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/22/2019] [Indexed: 12/26/2022] Open
Abstract
Therapies that target the function of immune cells have significant clinical efficacy in diseases such as cancer and autoimmunity. Although functional genomics has accelerated therapeutic target discovery in cancer, its use in primary immune cells is limited because vector delivery is inefficient and can perturb cell states. Here we describe CHIME: CHimeric IMmune Editing, a CRISPR-Cas9 bone marrow delivery system to rapidly evaluate gene function in innate and adaptive immune cells in vivo without ex vivo manipulation of these mature lineages. This approach enables efficient deletion of genes of interest in major immune lineages without altering their development or function. We use this approach to perform an in vivo pooled genetic screen and identify Ptpn2 as a negative regulator of CD8+ T cell-mediated responses to LCMV Clone 13 viral infection. These findings indicate that this genetic platform can enable rapid target discovery through pooled screening in immune cells in vivo.
Collapse
Affiliation(s)
- Martin W LaFleur
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Thao H Nguyen
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Matthew A Coxe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Kathleen B Yates
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Justin D Trombley
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Sarah A Weiss
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Flavian D Brown
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Jacob E Gillis
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Daniel J Coxe
- School of Energy, Matter, and Transport Engineering at Arizona State University, Tempe, AZ, 85287, USA
| | - John G Doench
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA.
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02142, USA.
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA.
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02142, USA.
| |
Collapse
|
39
|
Li Y, Zhou H, Li Y, Han L, Song M, Chen F, Shang G, Wang D, Wang Z, Zhang W, Zhong M. PTPN2 improved renal injury and fibrosis by suppressing STAT-induced inflammation in early diabetic nephropathy. J Cell Mol Med 2019; 23:4179-4195. [PMID: 30955247 PMCID: PMC6533506 DOI: 10.1111/jcmm.14304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 02/20/2019] [Accepted: 03/14/2019] [Indexed: 12/11/2022] Open
Abstract
Diabetic nephropathy (DN) is a chronic inflammatory disease triggered by disordered metabolism. Recent studies suggested that protein tyrosine phosphatase non‐receptor type 2 (PTPN2) could ameliorate metabolic disorders and suppress inflammatory responses. This study investigated PTPN2's role in modulating DN and the possible cellular mechanisms involved. In a mouse model combining hyperglycaemia and hypercholesterolaemia (streptozotocin diabetic, ApoE‐/‐ mice), mice showed severe insulin resistance, renal dysfunction, micro‐inflammation, subsequent extracellular matrix expansion and decreased expression of PTPN2. We found that mice treated with PTPN2 displayed reduced serum creatinine, serum BUN and proteinuria. PTPN2 gene therapy markedly attenuated metabolic disorders and hyperglycaemia. In addition, PTPN2 gene transfer significantly suppressed renal activation of signal transducers and activators of transcription (STAT), STAT‐dependent pro‐inflammatory and pro‐fibrotic genes expression, and influx of lymphocytes in DN, indicating anti‐inflammatory effects of PTPN2 by inhibiting the activation of STAT signalling pathway in vivo. Furthermore, PTPN2 overexpression inhibited the high‐glucose induced phosphorylation of STAT, target genes expression and proliferation in mouse mesangial and tubuloepithelial cells, suggesting that the roles of PTPN2 on STAT activation was independent of glycaemic changes. Our results demonstrated that PTPN2 gene therapy could exert protective effects on DN via ameliorating metabolic disorders and inhibiting renal STAT‐dependent micro‐inflammation, suggesting its potential role for treatment of human DN.
Collapse
Affiliation(s)
- Ya Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Huimin Zhou
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yulin Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lu Han
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of General Practice, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ming Song
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fangfang Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Guokai Shang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Di Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhihao Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of Geriatric Medicine, Qilu Hospital of Shandong University, Key Laboratory of Cardiovascular Proteomics of Shandong Province, Ji'nan, China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
40
|
Activation of naïve CD4 + T cells re-tunes STAT1 signaling to deliver unique cytokine responses in memory CD4 + T cells. Nat Immunol 2019; 20:458-470. [PMID: 30890796 PMCID: PMC7610646 DOI: 10.1038/s41590-019-0350-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/14/2019] [Indexed: 12/31/2022]
Abstract
The cytokine IL-6 controls the survival, proliferation and effector characteristics of lymphocytes through activation of the transcription factors STAT1 and STAT3. While STAT3 activity is an ever-present feature of IL-6 signaling in CD4+ T cells, prior T-cell receptor activation limits the IL-6 control of STAT1 in effector and memory populations. Here we show that STAT1 phosphorylation in response to IL-6 was regulated by protein tyrosine phosphatases (PTPN2, PTPN22) expressed in response to the activation of naïve CD4+ T cells. Transcriptomic and chromatin immunoprecipitation-sequencing of IL-6 responses in naïve and effector memory CD4+ T cells showed how the suppression of STAT1 activation shaped the functional identity and effector characteristics of memory CD4+ T cells. Thus, protein tyrosine phosphatases induced by activation of naïve T cells determined the way activated or memory CD4+ T cells sensed and interpreted cytokine signals.
Collapse
|
41
|
Hu XX, Wu YJ, Zhang J, Wei W. T-cells interact with B cells, dendritic cells, and fibroblast-like synoviocytes as hub-like key cells in rheumatoid arthritis. Int Immunopharmacol 2019; 70:428-434. [PMID: 30856393 DOI: 10.1016/j.intimp.2019.03.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/15/2019] [Accepted: 03/05/2019] [Indexed: 12/19/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory synovitis-based systemic disease characterized by invasive joint inflammation and synovial hyperplasia, which can lead to arthrentasis and defunctionalization. Previous research has shown that T cells, B cells, dendritic cells (DCs), and fibroblast-like synoviocytes (FLSs) play vital roles in the regulation of RA. Both T follicular helper (Tfh) cells and helper T (Th) 17 cells play immunomodulatory roles in RA. Moreover, interleukin-23 (IL-23), and IL-17 are vital to the pathogenesis of RA. T cells behave as a hub, in that B cells, DCs, and FLSs can interact with T cells to inhibit their activation and interfere with the process of RA. T cells cooperate with B cells, DCs, and FLSs to maintain the stability of the immune system under physiological conditions. However, under pathological conditions, the balance is disrupted, and the interaction of T cells with other cells may intensify disease progression. This review focuses on the interaction of T cells with B cells, DCs, and FLSs in different tissues and organs of RA patients and animal models, and highlight that the interplay between immune cells may underline the unique function of T cells and the application prospect of targeting T cell treatment for RA.
Collapse
Affiliation(s)
- Xiao-Xi Hu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Yu-Jing Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Jing Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China.
| |
Collapse
|
42
|
Svensson MN, Doody KM, Schmiedel BJ, Bhattacharyya S, Panwar B, Wiede F, Yang S, Santelli E, Wu DJ, Sacchetti C, Gujar R, Seumois G, Kiosses WB, Aubry I, Kim G, Mydel P, Sakaguchi S, Kronenberg M, Tiganis T, Tremblay ML, Ay F, Vijayanand P, Bottini N. Reduced expression of phosphatase PTPN2 promotes pathogenic conversion of Tregs in autoimmunity. J Clin Invest 2019; 129:1193-1210. [PMID: 30620725 DOI: 10.1172/jci123267] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 01/03/2019] [Indexed: 12/29/2022] Open
Abstract
Genetic variants at the PTPN2 locus, which encodes the tyrosine phosphatase PTPN2, cause reduced gene expression and are linked to rheumatoid arthritis (RA) and other autoimmune diseases. PTPN2 inhibits signaling through the T cell and cytokine receptors, and loss of PTPN2 promotes T cell expansion and CD4- and CD8-driven autoimmunity. However, it remains unknown whether loss of PTPN2 in FoxP3+ regulatory T cells (Tregs) plays a role in autoimmunity. Here we aimed to model human autoimmune-predisposing PTPN2 variants, the presence of which results in a partial loss of PTPN2 expression, in mouse models of RA. We identified that reduced expression of Ptpn2 enhanced the severity of autoimmune arthritis in the T cell-dependent SKG mouse model and demonstrated that this phenotype was mediated through a Treg-intrinsic mechanism. Mechanistically, we found that through dephosphorylation of STAT3, PTPN2 inhibits IL-6-driven pathogenic loss of FoxP3 after Tregs have acquired RORγt expression, at a stage when chromatin accessibility for STAT3-targeted IL-17-associated transcription factors is maximized. We conclude that PTPN2 promotes FoxP3 stability in mouse RORγt+ Tregs and that loss of function of PTPN2 in Tregs contributes to the association between PTPN2 and autoimmunity.
Collapse
Affiliation(s)
- Mattias Nd Svensson
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, and
| | | | - Benjamin J Schmiedel
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Sourya Bhattacharyya
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Bharat Panwar
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Florian Wiede
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Shen Yang
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Eugenio Santelli
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Dennis J Wu
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, and
| | - Cristiano Sacchetti
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, and
| | - Ravindra Gujar
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Gregory Seumois
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - William B Kiosses
- Core Microscopy, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Isabelle Aubry
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Gisen Kim
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Piotr Mydel
- Clinical Science, Broegelmann Research Laboratory, Bergen, Norway.,Department of Microbiology, Jagiellonian University, Krakow, Poland
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Tony Tiganis
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Monash Biomedicine Discovery Institute, and.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Michel L Tremblay
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Ferhat Ay
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Pandurangan Vijayanand
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Nunzio Bottini
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, and
| |
Collapse
|
43
|
Li J, Wu G, Cao Y, Hou Z. Roles of miR-210 in the pathogenesis of pre-eclampsia. Arch Med Sci 2019; 15:183-190. [PMID: 30697269 PMCID: PMC6348360 DOI: 10.5114/aoms.2018.73129] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION This study aimed to explore the bio-function of miR-210 in the pathogenesis of pre-eclampsia and provide new insights into the diagnosis and treatment of pre-eclampsia. MATERIAL AND METHODS A JAR cell line cultured in standard or hypoxic conditions was used in this study. Expression levels of miR-210 and PTPN2 were determined using real-time polymerase chain reaction (RT-PCR). Protein and phosphorylation levels were assessed using western blotting. Proliferation of JAR cells was evaluated using MTT assay. Migration and invasion were measured using transwell assay. RESULTS Expression of miR-210 increased significantly in a time-dependent manner after hypoxia treatment within 36 h (p < 0.05). miR-210 inhibitor significantly decreased the cell proliferation, migration, and invasion (p < 0.05), while miR-210 mimic reversed these findings (p < 0.05). Hypoxia significantly suppressed the expression of PTPN2; however, this elevation was abolished by miR-210 inhibitor (p < 0.05). Inhibition of PTPN2 or hypoxia significantly increased the proliferation, migration, and invasion of JAR cells, while miR-210 inhibitor significantly reversed these changes (p < 0.05). The phosphorylation levels of PDGFR, Akt, and Erk were markedly upregulated by hypoxia or si-PTPN2, but this effect was abolished by miR-210 inhibitor (p < 0.05). CONCLUSIONS miR-210 can promote proliferation, migration, and invasion via downregulating PTPN2 in the PDGFR-Akt pathway.
Collapse
Affiliation(s)
- Jiyun Li
- Third Department of Obstetrical, Hebei Cangzhou Central Hospital, Hebei, China
| | - Guimei Wu
- Third Department of Obstetrical, Hebei Cangzhou Central Hospital, Hebei, China
| | - Yanmin Cao
- Third Department of Obstetrical, Hebei Cangzhou Central Hospital, Hebei, China
| | - Zhi Hou
- Third Department of Obstetrical, Hebei Cangzhou Central Hospital, Hebei, China
| |
Collapse
|
44
|
Chen CP, Lin SP, Chern SR, Wu PS, Chen SW, Lai ST, Chuang TY, Chen WL, Wang W. A 13-year-old girl with 18p deletion syndrome presenting Turner syndrome-like clinical features of short stature, short webbed neck, low posterior hair line, puffy eyelids and increased carrying angle of the elbows. Taiwan J Obstet Gynecol 2018; 57:583-587. [PMID: 30122583 DOI: 10.1016/j.tjog.2018.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE We report a 13-year-old girl with 18p deletion syndrome presenting Turner syndrome-like clinical features. CASE REPORT A 13-year-old girl was referred for genetic counseling of Turner syndrome-like clinical features of short stature, short webbed neck, low posterior hair line, puffy eyelids and increased carrying angle of the elbows. The girl also had mild intellectual disability, psychomotor developmental delay, speech disorder, high-arched palate, hypertelorism and mid-face hypoplasia. Cytogenetic analysis of the girl revealed a karyotype of 46,XX,del(18) (p11.2). The parental karyotypes were normal. Array comparative genomic hybridization analysis on the DNA extracted from the peripheral blood revealed a 13.93-Mb deletion of 18p11.32-p11.21 or arr 18p11.32p11.21 (148,993-14,081,858) × 1.0 [GRCh37 (hg19)] encompassing 52 Online Mendelian Inheritance in Man (OMIM) genes including USP14, TYMS, SMCHD1, TGIF1, LAMA1, TWSG1, GNAL and PTPN2. Polymorphic DNA marker analysis revealed a maternal origin of the deletion. CONCLUSION Females with Turner syndrome-like clinical features in association with intellectual disability, facial dysmorphism and psychomotor developmental delay should be suspected of having chromosome deletion syndromes.
Collapse
Affiliation(s)
- Chih-Ping Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Institute of Clinical and Community Health Nursing, National Yang-Ming University, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Shuan-Pei Lin
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Department of Early Childhood Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Schu-Rern Chern
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | | | - Shin-Wen Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Shih-Ting Lai
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tzu-Yun Chuang
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Wen-Lin Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Wayseen Wang
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; Department of Bioengineering, Tatung University, Taipei, Taiwan
| |
Collapse
|
45
|
Rojas M, Restrepo-Jiménez P, Monsalve DM, Pacheco Y, Acosta-Ampudia Y, Ramírez-Santana C, Leung PS, Ansari AA, Gershwin ME, Anaya JM. Molecular mimicry and autoimmunity. J Autoimmun 2018; 95:100-123. [DOI: 10.1016/j.jaut.2018.10.012] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/12/2018] [Accepted: 10/16/2018] [Indexed: 12/15/2022]
|
46
|
Petersone L, Edner NM, Ovcinnikovs V, Heuts F, Ross EM, Ntavli E, Wang CJ, Walker LSK. T Cell/B Cell Collaboration and Autoimmunity: An Intimate Relationship. Front Immunol 2018; 9:1941. [PMID: 30210496 PMCID: PMC6119692 DOI: 10.3389/fimmu.2018.01941] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/06/2018] [Indexed: 12/17/2022] Open
Abstract
Co-ordinated interaction between distinct cell types is a hallmark of successful immune function. A striking example of this is the carefully orchestrated cooperation between helper T cells and B cells that occurs during the initiation and fine-tuning of T-cell dependent antibody responses. While these processes have evolved to permit rapid immune defense against infection, it is becoming increasingly clear that such interactions can also underpin the development of autoimmunity. Here we discuss a selection of cellular and molecular pathways that mediate T cell/B cell collaboration and highlight how in vivo models and genome wide association studies link them with autoimmune disease. In particular, we emphasize how CTLA-4-mediated regulation of CD28 signaling controls the engagement of secondary costimulatory pathways such as ICOS and OX40, and profoundly influences the capacity of T cells to provide B cell help. While our molecular understanding of the co-operation between T cells and B cells derives from analysis of secondary lymphoid tissues, emerging evidence suggests that subtly different rules may govern the interaction of T and B cells at ectopic sites during autoimmune inflammation. Accordingly, the phenotype of the T cells providing help at these sites includes notable distinctions, despite sharing core features with T cells imparting help in secondary lymphoid tissues. Finally, we highlight the interdependence of T cell and B cell responses and suggest that a significant beneficial impact of B cell depletion in autoimmune settings may be its detrimental effect on T cells engaged in molecular conversation with B cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lucy S. K. Walker
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|
47
|
Wang PF, Cai HQ, Zhang CB, Li YM, Liu X, Wan JH, Jiang T, Li SW, Yan CX. Molecular and clinical characterization of PTPN2 expression from RNA-seq data of 996 brain gliomas. J Neuroinflammation 2018; 15:145. [PMID: 29764444 PMCID: PMC5953404 DOI: 10.1186/s12974-018-1187-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/03/2018] [Indexed: 12/14/2022] Open
Abstract
Background Immune checkpoint inhibitors have been shown to promote antitumor immunity and achieve durable tumor remissions. However, certain tumors are refractory to current immunotherapy. These negative results encouraged us to uncover other therapeutic targets and strategies. PTPN2 (protein tyrosine phosphatase, non-receptor type 2) has been newly identified as an immunotherapy target. Loss of PTPN2 sensitizes the tumor to immunotherapy via IFNγ signaling. Methods Here, we investigated the relationship between PTPN2 mRNA levels and clinical characteristics in gliomas. RNA-seq data of a cohort of 325 patients with glioma were available from the Chinese Glioma Genome Atlas and 671 from The Cancer Genome Atlas. R language, GraphPad Prism 5, and SPSS 22.0 were used to analyze data and draw figures. Results PTPN2 transcript levels increased significantly with higher grades of glioma and in isocitrate dehydrogenase (IDH) wild-type and mesenchymal subtype gliomas. A comprehensive biological analysis was conducted, which indicated a crucial role of PTPN2 in the immune and inflammation responses in gliomas. Specifically, PTPN2 was positively associated with HCK, LCK, MHC II, and STAT1 but negatively related to IgG and interferon. Moreover, canonical correlation analysis showed a positive correlation of PTPN2 with infiltrating immune cells, such as macrophages, neutrophils, and CD8+ T cells. Clinically, higher levels of PTPN2 were associated with a worse overall survival both in patients with gliomas and glioblastomas. Conclusion PTPN2 expression level was increased in glioblastomas and associated with gliomas of the IDH wild-type and mesenchymal subtype. There was a close correlation between PTPN2 and the immune response and inflammatory activity in gliomas. Our results show that PTPN2 is a promising immunotherapy target and may provide additional treatment strategies. Electronic supplementary material The online version of this article (10.1186/s12974-018-1187-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Building 1, Ward 6, Xiang Shan Yi Ke Song Road 50, Haidian, Beijing, China
| | - Hong-Qing Cai
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Building 1, Ward 6, Xiang Shan Yi Ke Song Road 50, Haidian, Beijing, China.,Department of Neurosurgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuan-Bao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Neurosurgical Institute, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Yan-Michael Li
- Department of Neurosurgery and Oncology, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiang Liu
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Jing-Hai Wan
- Department of Neurosurgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Neurosurgical Institute, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Shou-Wei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Building 1, Ward 6, Xiang Shan Yi Ke Song Road 50, Haidian, Beijing, China.
| | - Chang-Xiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Building 1, Ward 6, Xiang Shan Yi Ke Song Road 50, Haidian, Beijing, China.
| |
Collapse
|
48
|
de Almeida RC, Chagas VS, Castro MAA, Petzl-Erler ML. Integrative Analysis Identifies Genetic Variants Associated With Autoimmune Diseases Affecting Putative MicroRNA Binding Sites. Front Genet 2018; 9:139. [PMID: 29755505 PMCID: PMC5932181 DOI: 10.3389/fgene.2018.00139] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/04/2018] [Indexed: 12/20/2022] Open
Abstract
Genome-wide and fine mapping studies have shown that more than 90% of genetic variants associated with autoimmune diseases (AID) are located in non-coding regions of the human genome and especially in regulatory sequences, including microRNAs (miRNA) target sites. MiRNAs are small endogenous noncoding RNAs that modulate gene expression at the post-transcriptional level. Single nucleotide polymorphisms (SNPs) located within the 3' untranslated region of their target mRNAs (miRSNP) can alter miRNA binding sites. Yet, little is known about their effect on regulation by miRNA and the consequences for AID. Conversely, it is well known that two or more AID may share part of their genetic background. Here, we hypothesized that miRSNPs could be associated with more than one AID. To identify miRSNPs associated with AID, we integrated results from three different prediction tools (Polymirts, miRSNP, and miRSNPscore) using a naïve Bayes classifier approach to identify miRSNPs predicted to affect binding sites of miRNAs. Further, to detect miRSNPs associated with two or more AID, we integrated predictions with summary statistics from 12 AID studies. In addition, to prioritize miRSNPs, miRNAs and AID-associated target genes, we used public expression quantitative trait locus (eQTL) data and mRNA-seq and small RNA-seq data. We identified 34 miRNSPs associated with at least two AID. Furthermore, we found 86 miRNAs predicted to target 18 of the associated gene's mRNAs. Our integrative approach revealed new insights into miRNAs and AID associated target genes. Thus, it helped to prioritize AID noncoding risk SNPs that might be involved in the causal mechanisms, providing valuable information for further functional studies.
Collapse
Affiliation(s)
- Rodrigo C. de Almeida
- Human Molecular Genetics Laboratory, Department of Genetics, Federal University of Paraná, Curitiba, Brazil
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| | - Vinícius S. Chagas
- Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba, Brazil
| | - Mauro A. A. Castro
- Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba, Brazil
| | - Maria L. Petzl-Erler
- Human Molecular Genetics Laboratory, Department of Genetics, Federal University of Paraná, Curitiba, Brazil
| |
Collapse
|
49
|
Okuno M, Ayabe T, Yokota I, Musha I, Shiga K, Kikuchi T, Kikuchi N, Ohtake A, Nakamura A, Nakabayashi K, Okamura K, Momozawa Y, Kubo M, Suzuki J, Urakami T, Kawamura T, Amemiya S, Ogata T, Sugihara S, Fukami M. Protein-altering variants of PTPN2 in childhood-onset Type 1A diabetes. Diabet Med 2018; 35:376-380. [PMID: 29247561 DOI: 10.1111/dme.13566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2017] [Indexed: 12/31/2022]
Abstract
AIM To examine the contribution of PTPN2 coding variants to the risk of childhood-onset Type 1A diabetes. METHODS PTPN2 mutation analysis was carried out for 169 unrelated Japanese people with childhood-onset Type 1A diabetes. We searched for coding variants that were absent or extremely rare in the general population and were scored as damaging by multiple in silico programs. We performed mRNA analysis and three-dimensional structural prediction of the detected variants, when possible. We also examined possible physical links between these variants and previously reported risk SNPs as well as clinical information from variant-positive children. RESULTS One frameshift variant (p.Q286Yfs*24) and two probably damaging missense substitutions (p.C232W and p.R350Q) were identified in one child each. Of these, p.Q286Yfs*24 and p.C232W were hitherto unreported, while p.R350Q accounted for 2/121,122 alleles of the exome datasets. The p.Q286Yfs*24 variant did not encode stable mRNA, and p.C232W appeared to affect the structure of the tyrosine-protein phosphatase domain. The three variants were physically unrelated to known risk SNPs. The variant-positive children manifested Type 1A diabetes without additional clinical features and invariably carried risk human leukocyte antigen alleles. CONCLUSIONS The results provide the first indication that PTPN2 variants contribute to the risk of Type 1A diabetes, independently of known risk SNPs. PTPN2 coding variants possibly induce non-specific Type 1A diabetes phenotypes in individuals with human leukocyte antigen-mediated disease susceptibility. Our findings warrant further validation.
Collapse
Affiliation(s)
- M Okuno
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo
- Department of Paediatrics and Child Health, Nihon University School of Medicine, Tokyo
| | - T Ayabe
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo
| | - I Yokota
- Department of Paediatrics, Division of Paediatric Endocrinology and Metabolism, Shikoku Medical Centre for Children and Adults, Kagawa
| | - I Musha
- Department of Paediatrics, Saitama Medical University, Faculty of Medicine, Saitama
| | - K Shiga
- Department of Paediatrics, Children's Medical Centre, Yokohama City University Medical Centre, Yokohama
| | - T Kikuchi
- Department of Paediatrics, Saitama Medical University, Faculty of Medicine, Saitama
| | - N Kikuchi
- Department of Paediatrics, Yokohama City Minato Red Cross Hospital, Yokohama
| | - A Ohtake
- Department of Paediatrics, Saitama Medical University, Faculty of Medicine, Saitama
| | - A Nakamura
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo
| | - K Nakabayashi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo
| | - K Okamura
- Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo
| | - Y Momozawa
- Laboratory for Genotyping Development, Riken Centre for Integrative Medical Sciences, Kanagawa
| | - M Kubo
- Laboratory for Genotyping Development, Riken Centre for Integrative Medical Sciences, Kanagawa
| | - J Suzuki
- Department of Paediatrics and Child Health, Nihon University School of Medicine, Tokyo
| | - T Urakami
- Department of Paediatrics and Child Health, Nihon University School of Medicine, Tokyo
| | - T Kawamura
- Department of Paediatrics, Osaka City University School of Medicine, Osaka
| | - S Amemiya
- Department of Paediatrics, Saitama Medical University, Faculty of Medicine, Saitama
| | - T Ogata
- Department of Paediatrics, Hamamatsu University School of Medicine, Hamamatsu
| | - S Sugihara
- Department of Paediatrics, Tokyo Women's Medical University Medical Centre East, Tokyo, Japan
| | - M Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo
| |
Collapse
|
50
|
Houtman M, Shchetynsky K, Chemin K, Hensvold AH, Ramsköld D, Tandre K, Eloranta ML, Rönnblom L, Uebe S, Catrina AI, Malmström V, Padyukov L. T cells are influenced by a long non-coding RNA in the autoimmune associated PTPN2 locus. J Autoimmun 2018; 90:28-38. [PMID: 29398253 DOI: 10.1016/j.jaut.2018.01.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/10/2018] [Accepted: 01/11/2018] [Indexed: 12/31/2022]
Abstract
Non-coding SNPs in the protein tyrosine phosphatase non-receptor type 2 (PTPN2) locus have been linked with several autoimmune diseases, including rheumatoid arthritis, type I diabetes, and inflammatory bowel disease. However, the functional consequences of these SNPs are poorly characterized. Herein, we show in blood cells that SNPs in the PTPN2 locus are highly correlated with DNA methylation levels at four CpG sites downstream of PTPN2 and expression levels of the long non-coding RNA (lncRNA) LINC01882 downstream of these CpG sites. We observed that LINC01882 is mainly expressed in T cells and that anti-CD3/CD28 activated naïve CD4+ T cells downregulate the expression of LINC01882. RNA sequencing analysis of LINC01882 knockdown in Jurkat T cells, using a combination of antisense oligonucleotides and RNA interference, revealed the upregulation of the transcription factor ZEB1 and kinase MAP2K4, both involved in IL-2 regulation. Overall, our data suggests the involvement of LINC01882 in T cell activation and hints towards an auxiliary role of these non-coding SNPs in autoimmunity associated with the PTPN2 locus.
Collapse
Affiliation(s)
- Miranda Houtman
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden.
| | - Klementy Shchetynsky
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Karine Chemin
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Aase Haj Hensvold
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Ramsköld
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Karolina Tandre
- Department of Medical Sciences, Rheumatology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Maija-Leena Eloranta
- Department of Medical Sciences, Rheumatology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Steffen Uebe
- Institute of Human Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Anca Irinel Catrina
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Vivianne Malmström
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Leonid Padyukov
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|