1
|
Kiani A, Pierotti CL, Schedel F, Kokot T, Weyershaeuser J, Brehm M, Rios P, Fehrenbach K, Warscheid B, Minguet S, Schamel WW, Köhn M. Development of a Peptide Inhibitor Targeting the C-SH2 Domain of the SHP2 Phosphatase. Chembiochem 2025; 26:e202400938. [PMID: 40318117 DOI: 10.1002/cbic.202400938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2025] [Revised: 05/02/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Src homology 2 (SH2) domain-containing phosphatase 2 (SHP2) mediates important signal transduction upon cell surface receptor stimulation, regulating multiple cellular functions. In addition to the catalytically active phosphotyrosine (pTyr) phosphatase domain, SHP2 contains two regulatory pTyr-binding domains: the N-SH2 and C-SH2 domains. While the role of the N-SH2 domain is well understood, the role of the C-SH2 domain is less clear. To support studies on the involvement of the domains in SHP2 function, herein, the development of a peptide inhibitor containing a nonhydrolysable pTyr mimetic, which selectively binds to the C-SH2 domain of SHP2 and blocks its protein-protein interactions, is described. Incorporation of the pTyr mimetic l-O-malonyltyrosine (l-OMT) results in robust binding affinity to the C-SH2 domain, while the widely used pTyr mimetic phosphonodifluoromethyl phenylalanine (F2Pmp) abolishes binding, showing that this mimetic is not a general binder of SH2 domains, which challenges existing notions. The C-SH2 inhibitor peptide (CSIP) is stable, selective, cell permeable, and noncytotoxic. CSIP enriches the toolbox of inhibitors with different modes of action targeting SHP2, and will support studies to better understand SHP2 regulation and interactions, which can ultimately inform new drug discovery efforts.
Collapse
Affiliation(s)
- Azin Kiani
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Faculty of Chemistry and Pharmacy, Hermann-Staudinger Graduate School, University of Freiburg, Hebelstraße 27, 79087, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
| | - Catia L Pierotti
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Käthe-Kümmel-Straße 1, 53115, Bonn, Germany
| | - Franziska Schedel
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Faculty of Chemistry and Pharmacy, Hermann-Staudinger Graduate School, University of Freiburg, Hebelstraße 27, 79087, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Käthe-Kümmel-Straße 1, 53115, Bonn, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstraße 19A, 79104, Freiburg im Breisgau, Germany
| | - Thomas Kokot
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
| | - Judith Weyershaeuser
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
| | - Mario Brehm
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
| | - Pablo Rios
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
| | - Kerstin Fehrenbach
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
| | - Bettina Warscheid
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Biochemistry II, Theodor-Boveri-Institute, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Susana Minguet
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
- Centre of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Breisacher Straße 115, 79106, Freiburg im Breisgau, Germany
| | - Wolfgang W Schamel
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
- Centre of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Breisacher Straße 115, 79106, Freiburg im Breisgau, Germany
| | - Maja Köhn
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg im Breisgau, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104, Freiburg im Breisgau, Germany
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Käthe-Kümmel-Straße 1, 53115, Bonn, Germany
| |
Collapse
|
2
|
Qi J, Cheng H, Su L, Li J, Cheng F. A novel exosome-related prognostic risk model for thyroid cancer. Asia Pac J Clin Oncol 2025; 21:190-198. [PMID: 38577908 DOI: 10.1111/ajco.14063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/13/2024] [Accepted: 03/20/2024] [Indexed: 04/06/2024]
Abstract
AIM The aim was to build an exosome-related gene (ERG) risk model for thyroid cancer (TC) patients. METHODS Note that, 510 TC samples from The Cancer Genome Atlas database and 121 ERGs from the ExoBCD database were obtained. Differential gene expression analysis was performed to get ERGs in TC (TERGs). Functional enrichment analyses including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were conducted on the TERGs. Then we constructed a model based on LASSO Cox regression analysis. Kaplan-Meier survival analysis was applied and a Nomogram model was also built. The immune landscape was evaluated by CIBERSORT. RESULTS Thirty-eight TERGs were identified and their functions were enriched on 591 GO terms and 30 KEGG pathways. We built a Risk Score model based on FGFR3, ADRA1B, and POSTN. Risk Scores were significantly higher in T4 than in other stages, meanwhile, it didn't significantly differ in genders and TNM N or M classifications. The nomogram model could reliably predict the overall survival of TC patients. The mutation rate of BRAF and expression of cytotoxic T-lymphocyte-associated protein 4 were significantly higher in the high-risk group than in the low-risk group. The risk score was significantly correlated to the immune landscape. CONCLUSION We built a Risk Score model using FGFR3, ADRA1B, and POSTN which could reliably predict the prognosis of TC patients.
Collapse
Affiliation(s)
- Junfeng Qi
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Hanshan Cheng
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Long Su
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Jun Li
- Department of Ultrasound, Wuwei People's Hospital, Wuwei, China
| | - Fei Cheng
- Department of Surgical Oncology, Wuwei People's Hospital, Wuwei, China
| |
Collapse
|
3
|
Mélique S, Vadel A, Rouquié N, Yang C, Bories C, Cotineau C, Saoudi A, Fazilleau N, Lesourne R. THEMIS promotes T cell development and maintenance by rising the signaling threshold of the inhibitory receptor BTLA. Proc Natl Acad Sci U S A 2024; 121:e2318773121. [PMID: 38713628 PMCID: PMC11098085 DOI: 10.1073/pnas.2318773121] [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: 10/26/2023] [Accepted: 04/12/2024] [Indexed: 05/09/2024] Open
Abstract
The current paradigm about the function of T cell immune checkpoints is that these receptors switch on inhibitory signals upon cognate ligand interaction. We here revisit this simple switch model and provide evidence that the T cell lineage protein THEMIS enhances the signaling threshold at which the immune checkpoint BTLA (B- and T-lymphocyte attenuator) represses T cell responses. THEMIS is recruited to the cytoplasmic domain of BTLA and blocks its signaling capacity by promoting/stabilizing the oxidation of the catalytic cysteine of the tyrosine phosphatase SHP-1. In contrast, THEMIS has no detectable effect on signaling pathways regulated by PD-1 (Programmed cell death protein 1), which depend mainly on the tyrosine phosphatase SHP-2. BTLA inhibitory signaling is tuned according to the THEMIS expression level, making CD8+ T cells more resistant to BTLA-mediated inhibition than CD4+ T cells. In the absence of THEMIS, the signaling capacity of BTLA is exacerbated, which results in the attenuation of signals driven by the T cell antigen receptor and by receptors for IL-2 and IL-15, consequently hampering thymocyte positive selection and peripheral CD8+ T cell maintenance. By characterizing the pivotal role of THEMIS in restricting the transmission of BTLA signals, our study suggests that immune checkpoint operability is conditioned by intracellular signal attenuators.
Collapse
Affiliation(s)
- Suzanne Mélique
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Aurélie Vadel
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Nelly Rouquié
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Cui Yang
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Cyrielle Bories
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Coline Cotineau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Abdelhadi Saoudi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Nicolas Fazilleau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Renaud Lesourne
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| |
Collapse
|
4
|
Hossen MM, Ma Y, Yin Z, Xia Y, Du J, Huang JY, Huang JJ, Zou L, Ye Z, Huang Z. Current understanding of CTLA-4: from mechanism to autoimmune diseases. Front Immunol 2023; 14:1198365. [PMID: 37497212 PMCID: PMC10367421 DOI: 10.3389/fimmu.2023.1198365] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023] Open
Abstract
Autoimmune diseases (ADs) are characterized by the production of autoreactive lymphocytes, immune responses to self-antigens, and inflammation in related tissues and organs. Cytotoxic T-lymphocyte antigen 4 (CTLA-4) is majorly expressed in activated T cells and works as a critical regulator in the inflammatory response. In this review, we first describe the structure, expression, and how the signaling pathways of CTLA-4 participate in reducing effector T-cell activity and enhancing the immunomodulatory ability of regulatory T (Treg) cells to reduce immune response, maintain immune homeostasis, and maintain autoimmune silence. We then focused on the correlation between CTLA-4 and different ADs and how this molecule regulates the immune activity of the diseases and inhibits the onset, progression, and pathology of various ADs. Finally, we summarized the current progress of CTLA-4 as a therapeutic target for various ADs.
Collapse
Affiliation(s)
- Md Munnaf Hossen
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Yanmei Ma
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhihua Yin
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Yuhao Xia
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jing Du
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jim Yi Huang
- Department of Psychology, University of Oklahoma, Norman, OK, United States
| | - Jennifer Jin Huang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Linghua Zou
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Rehabilitation Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhizhong Ye
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhong Huang
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| |
Collapse
|
5
|
Yang C, Li R, Su LC, Lan YY, Wang YQ, Xu WD, Huang AF. SHP2: its association and roles in systemic lupus erythematosus. Inflamm Res 2023:10.1007/s00011-023-01760-w. [PMID: 37351631 DOI: 10.1007/s00011-023-01760-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/04/2023] [Accepted: 06/17/2023] [Indexed: 06/24/2023] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is an autoimmune inflammatory disease. Src homology 2 domain containing protein tyrosine phosphatase (SHP2) is a member of the protein tyrosine phosphatases (PTPs) family. To date, relationship between SHP2 and SLE pathogenesis is not elucidated. METHOD We measured plasma levels of SHP2 in 328 SLE patients, 78 RA patients, 80 SS patients and 79 healthy controls by ELISA, and discussed association of SHP2 in SLE patients, potential of plasma SHP2 as a SLE biomarker. Moreover, histological and serological changes were evaluated by flow cytometry, HE/Masson examination, immunofluorescence test in pristane-induced lupus mice after SHP2 inhibitor injection to reveal role of SHP2 in lupus development. RESULTS Results indicated that SHP2 plasma levels were upregulated in SLE patients and correlated with some clinical, laboratory characteristics such as proteinuria, pyuria, and may be a potential biomarker for SLE. After SHP2 inhibitor treatment, hepatosplenomegaly and histological severity of the kidney in lupus mice were improved. SHP2 inhibitor reversed DCs, Th1, and Th17 cells differentiation and downregulated inflammatory cytokines (IL-4, IL-6, IL-10, IL-17A, IFN-γ and TNF-α) and autoantibodies (ANA, anti-dsDNA) production in pristane-lupus mice. CONCLUSION In summary, SHP2 correlated with SLE pathogenesis and promoted the development of lupus.
Collapse
Affiliation(s)
- Chan Yang
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, 646000, Sichuan, China
| | - Rong Li
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, 646000, Sichuan, China
| | - Lin-Chong Su
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, 2 Wufengshan Road, Enshi, 445000, Hubei, China
| | - You-Yu Lan
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, Sichuan, China
| | - You-Qiang Wang
- Department of Laboratory Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, 646000, Sichuan, China.
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
6
|
Wu M, Gao X, Tang Y, Wu W, Zhou J, Shao Y, Hao C, Yang Y, Zhang J. Cbl-b inhibited CD4 + T cell activation by regulating the expression of miR-99a/miR-125b. Int Immunopharmacol 2023; 115:109677. [PMID: 36634415 DOI: 10.1016/j.intimp.2022.109677] [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: 09/22/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023]
Abstract
The molecular regulation of T cell activation has always been a hot topic in immunology. It has been reported that Cbl-b inhibits T cell activation, but the specific molecular mechanism especially for transcriptional regulation has not been very clear so far. Our present study showed that ablation of Cbl-b resulted in the increased expression of miR-99a and miR-125b, and the antagonism of miR-99a or miR-125b could inhibit the Cbl-b-/- T cell over-activation partly. Further study demonstrated that Cbl-b could bind and ubiquitinate SHP-2 in the activated T cells. The activation of SHP-2 deficient T cells was significantly inhibited. Western blot showed that SHP-2 could dephosphorylate HOXA10, and HOXA10 could enter the nucleus under the stimulation of anti-CD3 antibody alone in Cbl-b deficient T cells. Luciferase reporter assay and CUT&Tag qPCR showed that HOXA10 could regulate the expression of miR-99a/miR-125b. Real-time PCR and western blot further indicated that miR-99a/miR-125b functioned on PI3K/AKT pathway to regulate T cell activation. In conclusion, our study demonstrated that Cbl-b ubiquitinated SHP-2 to arrest HOXA10-mediated CD4+ T cell activation by regulating the expression of miR-99a/miR-125b and their function on PI3K/AKT pathway, which might providing a new explanation for the regulation of T cell activation and potential new idea for autoimmune diseases and tumor immunotherapies.
Collapse
Affiliation(s)
- Mengyun Wu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Xiu Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yuxu Tang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Wenyan Wu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Ji Zhou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yu Shao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Chuangli Hao
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, People's Republic of China.
| | - Yi Yang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China.
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China.
| |
Collapse
|
7
|
Awadasseid A, Zhou Y, Zhang K, Tian K, Wu Y, Zhang W. Current studies and future promises of PD-1 signal inhibitors in cervical cancer therapy. Biomed Pharmacother 2023; 157:114057. [PMID: 36463828 DOI: 10.1016/j.biopha.2022.114057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/19/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
PD-1 (Programmed cell death-1) is a receptor that inhibits the activation of T cells and is an important target for cancer immunotherapy. PD-1 expression stays high on antigen-specific T cells that have been stimulated for a long time, making them less responsive to stimuli. Consequently, there has been a recent surge in the number of researchers focusing on how the PD-1 axis delivers inhibitory signals to uncover new therapeutic targets. As an inhibitory signaling mechanism, the PD-1 axis controls immunological responses. Blocking the PD-1 axis has been shown to have long-lasting effects on various cancers, demonstrating the crucial role of PD-1 in blocking anti-tumor immunity. Despite this role, most patients do not respond to PD-1 monotherapy, and some have experienced adverse events. Many challenges remain regarding the PD-1 signaling axis to be addressed. In this review, we outline the most recent research and prospects of PD-1 signal inhibitors to enhance cervical cancer therapy.
Collapse
Affiliation(s)
- Annoor Awadasseid
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Moganshan Institute ZJUT, Deqing 313202, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China; Department of Biochemistry & Food Sciences, University of Kordofan, El-Obeid 51111, Sudan
| | - Yongnan Zhou
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Koutian Zhang
- Zhejiang Jianing Pharmaceutical Technology Co., Ltd, Hangzhou 310051, China
| | - Kaiming Tian
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yanling Wu
- Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China.
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
8
|
Cammann C, Israel N, Frentzel S, Jeron A, Topfstedt E, Schüler T, Simeoni L, Zenker M, Fehling HJ, Schraven B, Bruder D, Seifert U. T cell-specific constitutive active SHP2 enhances T cell memory formation and reduces T cell activation. Front Immunol 2022; 13:958616. [PMID: 35983034 PMCID: PMC9379337 DOI: 10.3389/fimmu.2022.958616] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Upon antigen recognition by the T cell receptor (TCR), a complex signaling network orchestrated by protein-tyrosine kinases (PTKs) and protein-tyrosine phosphatases (PTPs) regulates the transmission of the extracellular signal to the nucleus. The role of the PTPs Src-homology 2 (SH2) domain-containing phosphatase 1 (SHP1, Ptpn6) and Src-homology 2 (SH2) domain-containing phosphatase 2 (SHP2, Ptpn11) have been studied in various cell types including T cells. Whereas SHP1 acts as an essential negative regulator of the proximal steps in T cell signalling, the role of SHP2 in T cell activation is still a matter of debate. Here, we analyzed the role of the constitutively active SHP2-D61Y-mutant in T cell activation using knock-in mice expressing the mutant form Ptpn11D61Y in T cells. We observed reduced numbers of CD8+ and increased numbers of CD4+ T cells in the bone marrow and spleen of young and aged SHP2-D61Y-mutant mice as well as in Influenza A Virus (IAV)-infected mice compared to controls. In addition, we found elevated frequencies of effector memory CD8+ T cells and an upregulation of the programmed cell death protein 1 (PD-1)-receptor on both CD4+ and CD8+ T cells. Functional analysis of SHP2-D61Y-mutated T cells revealed an induction of late apoptosis/necrosis, a reduced proliferation and altered signaling upon TCR stimulation. However, the ability of D61Y-mutant mice to clear viral infection was not affected. In conclusion, our data indicate an important regulatory role of SHP2 in T cell function, where the effect is determined by the kinetics of SHP2 phosphatase activity and differs in the presence of the permanently active and the temporally regulated phosphatase. Due to interaction of SHP2 with the PD-1-receptor targeting the protein-tyrosine phosphatase might be a valuable tool to enhance T cell activities in immunotherapy.
Collapse
Affiliation(s)
- Clemens Cammann
- Friedrich Loeffler-Institute for Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Nicole Israel
- Friedrich Loeffler-Institute for Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Sarah Frentzel
- Institute of Medical Microbiology, Infection Prevention and Control, Infection Immunology Group, Health Campus Immunology, Infectiology and Inflammation, Ottovon-Guericke-University Magdeburg, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Andreas Jeron
- Institute of Medical Microbiology, Infection Prevention and Control, Infection Immunology Group, Health Campus Immunology, Infectiology and Inflammation, Ottovon-Guericke-University Magdeburg, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Eylin Topfstedt
- Friedrich Loeffler-Institute for Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Luca Simeoni
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Dunja Bruder
- Institute of Medical Microbiology, Infection Prevention and Control, Infection Immunology Group, Health Campus Immunology, Infectiology and Inflammation, Ottovon-Guericke-University Magdeburg, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Ulrike Seifert
- Friedrich Loeffler-Institute for Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- *Correspondence: Ulrike Seifert,
| |
Collapse
|
9
|
Moore EK, Strazza M, Mor A. Combination Approaches to Target PD-1 Signaling in Cancer. Front Immunol 2022; 13:927265. [PMID: 35911672 PMCID: PMC9330480 DOI: 10.3389/fimmu.2022.927265] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer remains the second leading cause of death in the US, accounting for 25% of all deaths nationwide. Immunotherapy techniques bolster the immune cells' ability to target malignant cancer cells and have brought immense improvements in the field of cancer treatments. One important inhibitory protein in T cells, programmed cell death protein 1 (PD-1), has become an invaluable target for cancer immunotherapy. While anti-PD-1 antibody therapy is extremely successful in some patients, in others it fails or even causes further complications, including cancer hyper-progression and immune-related adverse events. Along with countless translational studies of the PD-1 signaling pathway, there are currently close to 5,000 clinical trials for antibodies against PD-1 and its ligand, PD-L1, around 80% of which investigate combinations with other therapies. Nevertheless, more work is needed to better understand the PD-1 signaling pathway and to facilitate new and improved evidence-based combination strategies. In this work, we consolidate recent discoveries of PD-1 signaling mediators and their therapeutic potential in combination with anti-PD-1/PD-L1 agents. We focus on the phosphatases SHP2 and PTPN2; the kinases ITK, VRK2, GSK-3, and CDK4/6; and the signaling adaptor protein PAG. We discuss their biology both in cancer cells and T cells, with a focus on their role in relation to PD-1 to determine their potential in therapeutic combinations. The literature discussed here was obtained from a search of the published literature and ClinicalTrials.gov with the following key terms: checkpoint inhibition, cancer immunotherapy, PD-1, PD-L1, SHP2, PTPN2, ITK, VRK2, CDK4/6, GSK-3, and PAG. Together, we find that all of these proteins are logical and promising targets for combination therapy, and that with a deeper mechanistic understanding they have potential to improve the response rate and decrease adverse events when thoughtfully used in combination with checkpoint inhibitors.
Collapse
Affiliation(s)
- Emily K. Moore
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| | - Marianne Strazza
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| | - Adam Mor
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
10
|
Yang C, Blaize G, Marrocco R, Rouquié N, Bories C, Gador M, Mélique S, Joulia E, Benamar M, Dejean AS, Daniels-Treffandier H, Love PE, Fazilleau N, Saoudi A, Lesourne R. THEMIS enhances the magnitude of normal and neuroinflammatory type 1 immune responses by promoting TCR-independent signals. Sci Signal 2022; 15:eabl5343. [DOI: 10.1126/scisignal.abl5343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Signals that determine the differentiation of naïve CD4
+
T helper (T
H
) cells into specific effector cell subsets are primarily stimulated by cytokines, but additional signals are required to adjust the magnitude of T
H
cell responses and set the balance between effective immunity and immunological tolerance. By inducing the post-thymic deletion of the T cell lineage signaling protein THEMIS, we showed that THEMIS promoted the development of optimal type 1 immune responses to foreign antigens but stimulated signals that favored encephalitogenic responses to self-neuroantigens. THEMIS was required to stimulate the expression of the gene encoding the transcriptional regulator T-BET and the production of the cytokine interferon-γ (IFN-γ), and it enhanced the ability of encephalitogenic CD4
+
T cells to migrate into the central nervous system. Consistently, analysis of THEMIS expression in polarized CD4
+
T cells showed that THEMIS was selectively increased in abundance in T
H
1 cells. The stimulation of predifferentiated effector CD4
+
T cells with antigen-presenting cells revealed a stimulatory function for THEMIS on type 1 cytokine responses, similar to those observed ex vivo after immunization. In contrast, THEMIS exerted opposing effects on naïve CD4
+
T cells in vitro by inhibiting the T cell receptor (TCR)–mediated signals that lead to T
H
1 cell responses. These data suggest that THEMIS exerts TCR-independent functions in effector T cells, which increase the magnitude of normal and pathogenic T
H
1 cell–mediated responses.
Collapse
Affiliation(s)
- Cui Yang
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Gaëtan Blaize
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Rémi Marrocco
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Nelly Rouquié
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Cyrielle Bories
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Mylène Gador
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Suzanne Mélique
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Emeline Joulia
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Mehdi Benamar
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Anne S. Dejean
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Hélène Daniels-Treffandier
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
- Ecole Nationale Vétérinaire de Toulouse, Toulouse, France
| | - Paul E. Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicolas Fazilleau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Abdelhadi Saoudi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Renaud Lesourne
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| |
Collapse
|
11
|
Immune Checkpoint Receptors Signaling in T Cells. Int J Mol Sci 2022; 23:ijms23073529. [PMID: 35408889 PMCID: PMC8999077 DOI: 10.3390/ijms23073529] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
The characterization of the receptors negatively modulating lymphocyte function is rapidly advancing, driven by success in tumor immunotherapy. As a result, the number of immune checkpoint receptors characterized from a functional perspective and targeted by innovative drugs continues to expand. This review focuses on the less explored area of the signaling mechanisms of these receptors, of those expressed in T cells. Studies conducted mainly on PD-1, CTLA-4, and BTLA have evidenced that the extracellular parts of some of the receptors act as decoy receptors for activating ligands, but in all instances, the tyrosine phosphorylation of their cytoplasmatic tail drives a crucial inhibitory signal. This negative signal is mediated by a few key signal transducers, such as tyrosine phosphatase, inositol phosphatase, and diacylglycerol kinase, which allows them to counteract TCR-mediated activation. The characterization of these signaling pathways is of great interest in the development of therapies for counteracting tumor-infiltrating lymphocyte exhaustion/anergy independently from the receptors involved.
Collapse
|
12
|
Mélique S, Yang C, Lesourne R. Negative times negative equals positive, THEMIS sets the rule on thymic selection and peripheral T cell responses. Biomed J 2022; 45:334-346. [PMID: 35346866 PMCID: PMC9250082 DOI: 10.1016/j.bj.2022.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/03/2022] [Accepted: 03/03/2022] [Indexed: 12/31/2022] Open
Abstract
The activity of T cells is finely controlled by a set of negative regulators of T-cell antigen receptor (TCR)-mediated signaling. However, how those negative regulators are themselves controlled to prevent ineffective TCR-mediated responses remain poorly understood. Thymocyte-expressed molecule involved in selection (THEMIS) has been characterized over a decade ago as an important player of T cell development. Although the molecular function of THEMIS has long remained puzzling and subject to controversies, latest investigations suggest that THEMIS stimulates TCR-mediated signaling by repressing the tyrosine phosphatases SHP-1 and SHP-2 which exert regulatory function on T cell activation. Recent evidences also point to a role for THEMIS in peripheral T cells beyond its role on thymic selection. Here, we present an overview of the past research on THEMIS in the context of T cell development and peripheral T cell function and discuss the possible implication of THEMIS-based mechanisms on TCR-dependent and independent signaling outcomes.
Collapse
Affiliation(s)
- Suzanne Mélique
- Infinity, University of Toulouse, CNRS5051, INSERM1291, UPS, Toulouse, France
| | - Cui Yang
- Infinity, University of Toulouse, CNRS5051, INSERM1291, UPS, Toulouse, France
| | - Renaud Lesourne
- Infinity, University of Toulouse, CNRS5051, INSERM1291, UPS, Toulouse, France.
| |
Collapse
|
13
|
Mor A, Strazza M. Bridging the Gap: Connecting the Mechanisms of Immune-Related Adverse Events and Autoimmunity Through PD-1. Front Cell Dev Biol 2022; 9:790386. [PMID: 35047501 PMCID: PMC8762228 DOI: 10.3389/fcell.2021.790386] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/30/2021] [Indexed: 12/19/2022] Open
Abstract
The emergence of anti-cytotoxic T-lymphocyte antigen 4 (anti-CTLA-4), anti-programmed cell death 1 ligand (anti-PD-1), and anti-PD-L1 antibodies as immune checkpoint inhibitors (ICIs) revolutionized the treatment of numerous types of tumors. These antibodies, both alone and in combination, provide great clinical efficacy as evidenced by tumor regression and increased overall patients' survival. However, with this success comes multiple challenges. First, while patients who respond to ICIs have outstanding outcomes, there remains a large proportion of patients who do not respond at all. This all-or-none response has led to looking downstream of programmed cell death 1 (PD-1) for additional therapeutic targets and for new combination therapies. Second, a majority of patients who receive ICIs go on to develop immune-related adverse events (irAEs) characterized by end-organ inflammation with T-cell infiltrates. The hallmarks of these clinically observed irAEs share many similarities with primary autoimmune diseases. The contribution of PD-1 to peripheral tolerance is a major mechanism for protection against expansion of self-reactive T-cell clones and autoimmune disease. In this review, we aim to bridge the gaps between our cellular and molecular knowledge of PD-1 signaling in T cells, ICI-induced irAEs, and autoimmune diseases. We will highlight shared mechanisms and the potential for new therapeutic strategies.
Collapse
Affiliation(s)
- Adam Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Marianne Strazza
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
14
|
Strazza M, Adam K, Lerrer S, Straube J, Sandigursky S, Ueberheide B, Mor A. SHP2 Targets ITK Downstream of PD-1 to Inhibit T Cell Function. Inflammation 2021; 44:1529-1539. [PMID: 33624224 PMCID: PMC9199348 DOI: 10.1007/s10753-021-01437-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/28/2021] [Accepted: 02/08/2021] [Indexed: 01/13/2023]
Abstract
PD-1 is a critical therapeutic target in cancer immunotherapy and antibodies blocking PD-1 are approved for multiple types of malignancies. The phosphatase SHP2 is the main effector mediating PD-1 downstream signaling and accordingly attempts have been made to target this enzyme as an alternative approach to treat immunogenic tumors. Unfortunately, small molecule inhibitors of SHP2 do not work as expected, suggesting that the role of SHP2 in T cells is more complex than initially hypothesized. To better understand the perplexing role of SHP2 in T cells, we performed interactome mapping of SAP, an adapter protein that is associated with SHP2 downstream signaling. Using genetic and pharmacological approaches, we discovered that SHP2 dephosphorylates ITK specifically downstream of PD-1 and that this event was associated with PD-1 inhibitory cellular functions. This study suggests that ITK is a unique target in this pathway, and since ITK is a SHP2-dependent specific mediator of PD-1 signaling, the combination of ITK inhibitors with PD-1 blockade may improve upon PD-1 monotherapy in the treatment of cancer.
Collapse
Affiliation(s)
- Marianne Strazza
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Kieran Adam
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Shalom Lerrer
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Johanna Straube
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Sabina Sandigursky
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Beatrix Ueberheide
- Proteomics Laboratory, New York University School of Medicine, New York, NY, 10016, USA
| | - Adam Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, 10032, USA.
- Division of Rheumatology, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
15
|
Patsoukis N, Wang Q, Strauss L, Boussiotis VA. Revisiting the PD-1 pathway. SCIENCE ADVANCES 2020; 6:6/38/eabd2712. [PMID: 32948597 PMCID: PMC7500922 DOI: 10.1126/sciadv.abd2712] [Citation(s) in RCA: 339] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/31/2020] [Indexed: 05/21/2023]
Abstract
Programmed Death-1 (PD-1; CD279) is an inhibitory receptor induced in activated T cells. PD-1 engagement by its ligands, PD-L1 and PD-L2, maintains peripheral tolerance but also compromises anti-tumor immunity. Blocking antibodies against PD-1 or its ligands have revolutionized cancer immunotherapy. However, only a fraction of patients develop durable antitumor responses. Clinical outcomes have reached a plateau without substantial advances by combinatorial approaches. Thus, great interest has recently emerged to investigate, in depth, the mechanisms by which the PD-1 pathway transmits inhibitory signals with the goal to identify molecular targets for improvement of the therapeutic success. These efforts have revealed unpredictable dimensions of the pathway and uncovered novel mechanisms involved in PD-1 and PD-L1 regulation and function. Here, we provide an overview of the recent advances on the mechanistic aspects of the PD-1 pathway and discuss the implications of these new discoveries and the gaps that remain to be filled.
Collapse
Affiliation(s)
- Nikolaos Patsoukis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Qi Wang
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Laura Strauss
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
16
|
The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat Immunol 2020; 21:1346-1358. [DOI: 10.1038/s41590-020-0769-3] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 07/28/2020] [Indexed: 02/06/2023]
|
17
|
Yuan Y, Fan Y, Gao Z, Sun X, Zhang H, Wang Z, Cui Y, Song W, Wang Z, Zhang F, Niu R. SHP2 promotes proliferation of breast cancer cells through regulating Cyclin D1 stability via the PI3K/AKT/GSK3β signaling pathway. Cancer Biol Med 2020; 17:707-725. [PMID: 32944401 PMCID: PMC7476086 DOI: 10.20892/j.issn.2095-3941.2020.0056] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/06/2020] [Indexed: 02/06/2023] Open
Abstract
Objective: The tyrosine phosphatase SHP2 has a dual role in cancer initiation and progression in a tissue type-dependent manner. Several studies have linked SHP2 to the aggressive behavior of breast cancer cells and poorer outcomes in people with cancer. Nevertheless, the mechanistic details of how SHP2 promotes breast cancer progression remain largely undefined. Methods: The relationship between SHP2 expression and the prognosis of patients with breast cancer was investigated by using the TCGA and GEO databases. The expression of SHP2 in breast cancer tissues was analyzed by immunohistochemistry. CRISPR/Cas9 technology was used to generate SHP2-knockout breast cancer cells. Cell-counting kit-8, colony formation, cell cycle, and EdU incorporation assays, as well as a tumor xenograft model were used to examine the function of SHP2 in breast cancer proliferation. Quantitative RT-PCR, western blotting, immunofluorescence staining, and ubiquitination assays were used to explore the molecular mechanism through which SHP2 regulates breast cancer proliferation. Results: High SHP2 expression is correlated with poor prognosis in patients with breast cancer. SHP2 is required for the proliferation of breast cancer cells in vitro and tumor growth in vivo through regulation of Cyclin D1 abundance, thereby accelerating cell cycle progression. Notably, SHP2 modulates the ubiquitin-proteasome-dependent degradation of Cyclin D1 via the PI3K/AKT/GSK3β signaling pathway. SHP2 knockout attenuates the activation of PI3K/AKT signaling and causes the dephosphorylation and resultant activation of GSK3β. GSK3β then mediates phosphorylation of Cyclin D1 at threonine 286, thereby promoting the translocation of Cyclin D1 from the nucleus to the cytoplasm and facilitating Cyclin D1 degradation through the ubiquitin-proteasome system. Conclusions: Our study uncovered the mechanism through which SHP2 regulates breast cancer proliferation. SHP2 may therefore potentially serve as a therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Yue Yuan
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Yanling Fan
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Zicong Gao
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Xuan Sun
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - He Zhang
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Zhiyong Wang
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Yanfen Cui
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Weijie Song
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Zhaosong Wang
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Fei Zhang
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Ruifang Niu
- Department of Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| |
Collapse
|
18
|
Niogret C, Birchmeier W, Guarda G. SHP-2 in Lymphocytes' Cytokine and Inhibitory Receptor Signaling. Front Immunol 2019; 10:2468. [PMID: 31708921 PMCID: PMC6823243 DOI: 10.3389/fimmu.2019.02468] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
Somewhat counterintuitively, the tyrosine phosphatase SHP-2 (SH2 domain-containing protein tyrosine phosphatase-2) is crucial for the activation of extracellular signal-regulated kinase (ERK) downstream of various growth factor receptors, thereby exerting essential developmental functions. This phosphatase also deploys proto-oncogenic functions and specific inhibitors have recently been developed. With respect to the immune system, the role of SHP-2 in the signaling of cytokines relevant for myelopoiesis and myeloid malignancies has been intensively studied. The function of this phosphatase downstream of cytokines important for lymphocytes is less understood, though multiple lines of evidence suggest its importance. In addition, SHP-2 has been proposed to mediate the suppressive effects of inhibitory receptors (IRs) that sustain a dysfunctional state in anticancer T cells. Molecules involved in IR signaling are of potential pharmaceutical interest as blockade of these inhibitory circuits leads to remarkable clinical benefit. Here, we discuss the dichotomy in the functions ascribed to SHP-2 downstream of cytokine receptors and IRs, with a focus on T and NK lymphocytes. Further, we highlight the importance of broadening our understanding of SHP-2′s relevance in lymphocytes, an essential step to inform on side effects and unanticipated benefits of its therapeutic blockade.
Collapse
Affiliation(s)
- Charlène Niogret
- Department of Biochemistry, University of Lausanne, Épalinges, Switzerland
| | - Walter Birchmeier
- Max-Delbrueck-Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| | - Greta Guarda
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
19
|
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
|
20
|
Zhao H, Martin E, Matalkah F, Shah N, Ivanov A, Ruppert JM, Lockman PR, Agazie YM. Conditional knockout of SHP2 in ErbB2 transgenic mice or inhibition in HER2-amplified breast cancer cell lines blocks oncogene expression and tumorigenesis. Oncogene 2019; 38:2275-2290. [PMID: 30467378 PMCID: PMC6440805 DOI: 10.1038/s41388-018-0574-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 09/13/2018] [Accepted: 10/04/2018] [Indexed: 01/21/2023]
Abstract
Overexpression of the human epidermal growth factor receptor 2 (HER2) is the cause of HER2-positive breast cancer (BC). Although HER2-inactivating therapies have benefited BC patients, development of resistance and disease recurrence have been the major clinical problems, pointing to a need for alternative therapeutic strategies. For that to happen, proteins that play critical roles in the biology of HER2-induced tumorigenesis have to be identified and characterized. Here, we show that the Src homology phosphotyrosyl phosphatase 2 (Shp2) encoded by the Ptpn11 gene is a requisite for ErbB2-induced tumorigenesis. We report that conditional knockout of Shp2 alleles in the ErbB2 BC model mice abrogates mammary tumorigenesis by blocking the expression of the ErbB2 transgene. We also show that inhibition of SHP2 encoded by the PTPN11 gene in the HER2-amplified BC cells induces a normal-like cellular phenotype and suppresses tumorigenesis and metastasis by blocking HER2 overexpression. These findings demonstrate that ErbB2-induced tumors in mice or xenograft tumors induced by transplantation of HER2-amplified BC cells are vulnerable to SHP2 inhibition since it abrogates the expression of the very oncogene that causes of the disease. This report paves the way for developing SHP2-targeting therapies for BC treatment in the future.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Elisha Martin
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Fatimah Matalkah
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Neal Shah
- Department of Basic Pharmaceutical Sciences, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Alexey Ivanov
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - J Michael Ruppert
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Paul R Lockman
- Department of Basic Pharmaceutical Sciences, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Yehenew M Agazie
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
21
|
Jiang Q. Natural forms of vitamin E and metabolites-regulation of cancer cell death and underlying mechanisms. IUBMB Life 2018; 71:495-506. [PMID: 30548200 DOI: 10.1002/iub.1978] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/17/2018] [Accepted: 11/02/2018] [Indexed: 12/25/2022]
Abstract
The disappointing results from large clinical studies of α-tocopherol (αT), the major form of vitamin E in tissues, for prevention of chronic diseases including cancer have cast doubt on not only αT but also other forms of vitamin E regarding their role in preventing carcinogenesis. However, basic research has shown that specific forms of vitamin E such as γ-tocopherol (γT), δ-tocopherol (δT), γ-tocotrienol (γTE) and δ-tocotrienol (δTE) can inhibit the growth and induce death of many types of cancer cells, and are capable of suppressing cancer development in preclinical cancer models. For these activities, these vitamin E forms are much stronger than αT. Further, recent research revealed novel anti-inflammatory and anticancer effects of vitamin E metabolites including 13'-carboxychromanols. This review focuses on anti-proliferation and induction of death in cancer cells by vitamin E forms and metabolites, and discuss mechanisms underlying these anticancer activities. The existing in vitro and in vivo evidence indicates that γT, δT, tocotrienols and 13'-carboxychromanols have anti-cancer activities via modulating key signaling or mediators that regulate cell death and tumor progression, such as eicosanoids, NF-κB, STAT3, PI3K, and sphingolipid metabolism. These results provide useful scientific rationales and mechanistic understanding for further translation of basic discoveries to the clinic with respect to potential use of these vitamin E forms and metabolites for cancer prevention and therapy. © 2018 IUBMB Life, 71(4):495-506, 2019.
Collapse
Affiliation(s)
- Qing Jiang
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, 47907, USA
| |
Collapse
|
22
|
Miah SMS, Jayasuriya CT, Salter AI, Reilly EC, Fugere C, Yang W, Chen Q, Brossay L. Ptpn11 Deletion in CD4 + Cells Does Not Affect T Cell Development and Functions but Causes Cartilage Tumors in a T Cell-Independent Manner. Front Immunol 2017; 8:1326. [PMID: 29085371 PMCID: PMC5650614 DOI: 10.3389/fimmu.2017.01326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 09/29/2017] [Indexed: 12/01/2022] Open
Abstract
The ubiquitously expressed tyrosine phosphatase Src homology region 2 domain-containing phosphatase-2 (SHP-2, encoded by Ptpn11) is required for constitutive cellular processes including proliferation, differentiation, and the regulation of immune responses. During development and maturation, subsets of T cells express a variety of inhibitory receptors known to associate with phosphatases, which in turn, dephosphorylate key players of activating receptor signaling pathways. We hypothesized that SHP-2 deletion would have major effects on T cell development by altering the thresholds for activation, as well as positive and negative selection. Surprisingly, using mice conditionally deficient for SHP-2 in the T cell lineage, we show that the development of these lymphocytes is globally intact. In addition, our data demonstrate that SHP-2 absence does not compromise T cell effector functions, suggesting that SHP-2 is dispensable in these cells. Unexpectedly, in aging mice, Ptpn11 gene deletion driven by CD4 Cre recombinase leads to cartilage tumors in wrist bones in a T cell-independent manner. These tumors resemble miniature cartilaginous growth plates and contain CD4-lineage positive chondrocyte-like cells. Altogether these results indicate that SHP-2 is a cartilage tumor suppressor during aging.
Collapse
Affiliation(s)
- S M Shahjahan Miah
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Rhode Island Hospital and Brown University Alpert Medical School, Providence, RI, United States
| | - Alexander I Salter
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| | - Emma C Reilly
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| | - Céline Fugere
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| | - Wentian Yang
- Department of Orthopaedics, Rhode Island Hospital and Brown University Alpert Medical School, Providence, RI, United States
| | - Qian Chen
- Department of Orthopaedics, Rhode Island Hospital and Brown University Alpert Medical School, Providence, RI, United States
| | - Laurent Brossay
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| |
Collapse
|
23
|
Mining the Complex Family of Protein Tyrosine Phosphatases for Checkpoint Regulators in Immunity. Curr Top Microbiol Immunol 2017; 410:191-214. [PMID: 28929190 DOI: 10.1007/82_2017_68] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The family of protein tyrosine phosphatases (PTPs) includes 107 genes in humans that are diverse in their structures and expression profiles. The majority are present in immune cells and play various roles in either inhibiting or promoting the duration and amplitude of signaling cascades. Several PTPs, including TC-PTP (PTPN2) and SHP-1 (PTPN6), have been recognized as being crucial for maintaining proper immune response and self-tolerance, and have gained recognition as true immune system checkpoint modulators. This chapter details the most recent literature on PTPs and immunity by examining their known functions in regulating signaling from either established checkpoint inhibitors or by their intrinsic properties, as modulators of the immune response. Notably, we review PTP regulatory properties in macrophages, antigen-presenting dendritic cells, and T cells. Overall, we present the PTP gene family as a remarkable source of novel checkpoint inhibitors wherein lies a great number of new targets for immunotherapies.
Collapse
|
24
|
Gu W, Prasadam I, Yu M, Zhang F, Ling P, Xiao Y, Yu C. Gamma tocotrienol targets tyrosine phosphatase SHP2 in mammospheres resulting in cell death through RAS/ERK pathway. BMC Cancer 2015; 15:609. [PMID: 26315028 PMCID: PMC4552156 DOI: 10.1186/s12885-015-1614-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/21/2015] [Indexed: 12/11/2022] Open
Abstract
Background There is increasing evidence supporting the concept of cancer stem cells (CSCs), which are responsible for the initiation, growth and metastasis of tumors. CSCs are thus considered the target for future cancer therapies. To achieve this goal, identifying potential therapeutic targets for CSCs is essential. Methods We used a natural product of vitamin E, gamma tocotrienol (gamma-T3), to treat mammospheres and spheres from colon and cervical cancers. Western blotting and real-time RT-PCR were employed to identify the gene and protein targets of gamma-T3 in mammospheres. Results We found that mammosphere growth was inhibited in a dose dependent manner, with total inhibition at high doses. Gamma-T3 also inhibited sphere growth in two other human epithelial cancers, colon and cervix. Our results suggested that both Src homology 2 domain-containing phosphatase 1 (SHP1) and 2 (SHP2) were affected by gamma-T3 which was accompanied by a decrease in K- and H-Ras gene expression and phosphorylated ERK protein levels in a dose dependent way. In contrast, expression of self-renewal genes TGF-beta and LIF, as well as ESR signal pathways were not affected by the treatment. These results suggest that gamma-T3 specifically targets SHP2 and the RAS/ERK signaling pathway. Conclusions SHP1 and SHP2 are potential therapeutic targets for breast CSCs and gamma-T3 is a promising natural drug for future breast cancer therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1614-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, The corner of Cooper Rd. St Lucia, Brisbane, QLD 4072, Australia.
| | - Indira Prasadam
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.
| | - Meihua Yu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, The corner of Cooper Rd. St Lucia, Brisbane, QLD 4072, Australia.
| | - Fengxia Zhang
- School of Biomedical Science, the University of Queensland, Brisbane, Australia.
| | - Patrick Ling
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, The corner of Cooper Rd. St Lucia, Brisbane, QLD 4072, Australia.
| |
Collapse
|
25
|
Clement M, Fornasa G, Loyau S, Morvan M, Andreata F, Guedj K, Khallou-Laschet J, Larghi P, Le Roux D, Bismuth G, Chiocchia G, Hivroz C, Newman DK, Nicoletti A, Caligiuri G. Upholding the T cell immune-regulatory function of CD31 inhibits the formation of T/B immunological synapses in vitro and attenuates the development of experimental autoimmune arthritis in vivo. J Autoimmun 2014; 56:23-33. [PMID: 25277651 DOI: 10.1016/j.jaut.2014.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 09/03/2014] [Accepted: 09/06/2014] [Indexed: 11/29/2022]
Abstract
CD31, a trans-homophilic inhibitory receptor expressed on both T- and B-lymphocytes, drives the mutual detachment of interacting leukocytes. Intriguingly, T cell CD31 molecules relocate to the immunological synapse (IS), where the T and B cells establish a stable interaction. Here, we show that intact CD31 molecules, which are able to drive an inhibitory signal, are concentrated at the periphery of the IS but are excluded from the center of the IS. At this site, were the cells establish the closest contact, the CD31 molecules are cleaved, and most of the extracellular portion of the protein, including the trans-homophilic binding sites, is shed from the cell surface. T cells lacking CD31 trans-homophilic binding sites easily establish stable interactions with B cells; at the opposite, CD31 signaling agonists inhibit T/B IS formation as well as the ensuing helper T cell activation and function. Confocal microscopy and flow cytometry analysis of experimental T/B IS shows that the T cell inhibitory effects of CD31 agonists depend on SHP-2 signaling, which reduces the phosphorylation of ZAP70. The analysis of synovial tissue biopsies from patients affected by rheumatoid arthritis showed that T cell CD31 molecules are excluded from the center of the T/B cell synapses in vivo. Interestingly, the administration of CD31 agonists in vivo significantly attenuated the development of the clinical signs of collagen-induced arthritis in DBA1/J mice. Altogether, our data indicate that the T cell co-inhibitory receptor CD31 prevents the formation of functional T/B immunological synapses and that therapeutic strategies aimed at sustaining CD31 signaling will attenuate the development of autoimmune responses in vivo.
Collapse
Affiliation(s)
- Marc Clement
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Giulia Fornasa
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Stéphane Loyau
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Marion Morvan
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Francesco Andreata
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Kevin Guedj
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Jamila Khallou-Laschet
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Paola Larghi
- Institut Curie, Section Recherche, Inserm U932, F-75005 Paris, France
| | - Delphine Le Roux
- Inserm U1016, Institut Cochin, F-75014 Paris, France; Centre National de la Recherche Scientifique, UMR8104, F-75014 Paris, France; Université Paris Descartes, Paris F-75014, France
| | - Georges Bismuth
- Inserm U1016, Institut Cochin, F-75014 Paris, France; Centre National de la Recherche Scientifique, UMR8104, F-75014 Paris, France; Université Paris Descartes, Paris F-75014, France
| | - Gilles Chiocchia
- Inserm U987, Faculté des Sciences de la Santé "Simone Veil", F-78180 Saint-Quentin-en-Yvelines, France; Laboratoire d'Excellence "Inflamex", F-75018 Paris, France; Université Versailles-Saint-Quentin, F-78180 Saint-Quentin-en-Yvelines, France
| | - Claire Hivroz
- Institut Curie, Section Recherche, Inserm U932, F-75005 Paris, France
| | - Debra K Newman
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53226, USA
| | - Antonino Nicoletti
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Giuseppina Caligiuri
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France.
| |
Collapse
|
26
|
Jang JY, Min JH, Chae YH, Baek JY, Wang SB, Park SJ, Oh GT, Lee SH, Ho YS, Chang TS. Reactive oxygen species play a critical role in collagen-induced platelet activation via SHP-2 oxidation. Antioxid Redox Signal 2014; 20:2528-40. [PMID: 24093153 PMCID: PMC4025609 DOI: 10.1089/ars.2013.5337] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS The collagen-stimulated generation of reactive oxygen species (ROS) regulates signal transduction in platelets, although the mechanism is unclear. The major targets of ROS include protein tyrosine phosphatases (PTPs). ROS-mediated oxidation of the active cysteine site in PTPs abrogates the PTP catalytic activity. The aim of this study was to elucidate whether collagen-induced ROS generation leads to PTP oxidation, which promotes platelet stimulation. RESULTS SH2 domain-containing PTP-2 (SHP-2) is oxidized in platelets by ROS produced upon collagen stimulation. The oxidative inactivation of SHP-2 leads to the enhanced tyrosine phosphorylation of spleen tyrosine kinase (Syk), Vav1, and Bruton's tyrosine kinase (Btk) in the linker for the activation of T cells signaling complex, which promotes the tyrosine phosphorylation-mediated activation of phospholipase Cγ2 (PLCγ2). Moreover, we found that, relative to wild-type platelets, platelets derived from glutathione peroxidase 1 (GPx1)/catalase double-deficient mice showed enhanced cellular ROS levels, oxidative inactivation of SHP-2, and tyrosine phosphorylation of Syk, Vav1, Btk, and PLCγ2 in response to collagen, which subsequently led to increased intracellular calcium levels, degranulation, and integrin αIIbβ3 activation. Consistent with these findings, GPx1/catalase double-deficiency accelerated the thrombotic response in FeCl3-injured carotid arteries. INNOVATION The present study is the first to demonstrate that SHP-2 is targeted by ROS produced in collagen-stimulated platelets and suggests that a novel mechanism for the regulation of platelet activation by ROS is due to oxidative inactivation of SHP-2. CONCLUSION We conclude that collagen-induced ROS production leads to SHP-2 oxidation, which promotes platelet activation by upregulating tyrosine phosphorylation-based signal transduction.
Collapse
Affiliation(s)
- Ji Yong Jang
- 1 Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University , Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hu Z, Fang H, Wang X, Chen D, Chen Z, Wang S. Overexpression of SHP2 tyrosine phosphatase promotes the tumorigenesis of breast carcinoma. Oncol Rep 2014; 32:205-12. [PMID: 24858400 DOI: 10.3892/or.2014.3201] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/30/2014] [Indexed: 11/05/2022] Open
Abstract
Expression of Src homology phosphotyrosine phosphatase 2 (SHP2) has been observed in human breast cancer. SHP2 is known to promote cell migration and invasiveness. However, the pathophysiologic role of SHP2 and its relevance to tumorigenesis are still largely unknown. In the present study, we aimed to evaluate the effect of SHP2 on the malignant phenotype of human breast cancer. An SHP2-overexpressing human breast cancer cell line was established by stable transfection of the SHP2 vector. The expression of SHP2 protein was detected using western blotting. The effects of SHP2 overexpression on cell proliferation were examined by an MTS assay. Invasion and migration abilities of the SHP2-overexpressing cells were determined using a Matrigel-based Boyden chamber invasion assay and a monolayer wound-healing assay. Increased SHP2 expression was detected following SHP2-vector transfection in the MDA-MB-231 cells. Overexpression of SHP2 was associated with increased cell proliferation and clone formation, and decreased chemotherapeutic sensitivity. Furthermore, transfection of SHP2 into breast cancer cells significantly promoted tumor growth in a mouse xenograft model. The mechanism of the promotion of tumorigenesis by SHP2 appears to involve its ability to increase the activity of ERK/AKT-mediated signaling pathways. In conclusion, our data suggest an important role of SHP2 in the molecular etiology of tumor growth, and implicate the potential application of SHP2 in cancer therapy.
Collapse
Affiliation(s)
- Zhongqian Hu
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Haoshu Fang
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xinyi Wang
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Danlei Chen
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhuo Chen
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Siying Wang
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
28
|
Rhee I, Veillette A. Protein tyrosine phosphatases in lymphocyte activation and autoimmunity. Nat Immunol 2012; 13:439-47. [PMID: 22513334 DOI: 10.1038/ni.2246] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lymphocyte activation must be tightly regulated to ensure sufficient immunity to pathogens and prevent autoimmunity. Protein tyrosine phosphatases (PTPs) serve critical roles in this regulation by controlling the functions of key receptors and intracellular signaling molecules in lymphocytes. In some cases, PTPs inhibit lymphocyte activation, whereas in others they promote it. Here we discuss recent progress in elucidating the roles and mechanisms of action of PTPs in lymphocyte activation. We also review the accumulating evidence that genetic alterations in PTPs are involved in human autoimmunity.
Collapse
Affiliation(s)
- Inmoo Rhee
- Laboratory of Molecular Oncology, Clinical Research Institute of Montréal, Montréal, Québec, Canada
| | | |
Collapse
|
29
|
Liu X, Qu CK. Protein Tyrosine Phosphatase SHP-2 (PTPN11) in Hematopoiesis and Leukemogenesis. JOURNAL OF SIGNAL TRANSDUCTION 2011; 2011:195239. [PMID: 21799948 PMCID: PMC3135119 DOI: 10.1155/2011/195239] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 04/01/2011] [Indexed: 01/28/2023]
Abstract
SHP-2 (PTPN11), a ubiquitously expressed protein tyrosine phosphatase, is critical for hematopoietic cell development and function owing to its essential role in growth factor/cytokine signaling. More importantly, germline and somatic mutations in this phosphatase are associated with Noonan syndrome, Leopard syndrome, and childhood hematologic malignancies. The molecular mechanisms by which SHP-2 mutations induce these diseases are not fully understood, as the biochemical bases of SHP-2 functions still remain elusive. Further understanding SHP-2 signaling activities and identification of its interacting proteins/substrates will shed light on the pathogenesis of PTPN11-associated hematologic malignancies, which, in turn, may lead to novel therapeutics for these diseases.
Collapse
Affiliation(s)
- Xia Liu
- Division of Hematology and Oncology, Department of Medicine, Center for Stem Cell and Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Cheng-Kui Qu
- Division of Hematology and Oncology, Department of Medicine, Center for Stem Cell and Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
30
|
Enhanced IL-12p40 production by phenylarsine oxide is mediated by cAMP response element in macrophages. Arch Pharm Res 2010; 33:745-51. [PMID: 20512473 DOI: 10.1007/s12272-010-0514-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 02/23/2010] [Accepted: 03/03/2010] [Indexed: 11/25/2022]
Abstract
Phenylarsine oxide (PAO), a membrane-permeable trivalent arsenical, is widely used as an inhibitor of protein tyrosine phosphatases. It reacts with vicinal sulfhydryl groups of proteins to form stable ring structures. Here we show the regulatory function of PAO in immune responses from macrophages. PAO significantly induced the secretion of interleukin (IL)-12p40 in lipopolysaccharide-stimulated macrophages. The mRNA expression and the gene promoter activity of IL-12p40 were enhanced by PAO. These results suggest that PAO may enhance IL-12p40 production at the transcriptional level. Furthermore, the effects of PAO on several signaling molecules regulating IL-12p40 expression were investigated. PAO attenuated the induced binding activity of cAMP response element (CRE), but not of NF-kappaB. Moreover, CRE promoter activity was dose-dependently inhibited by PAO and the increased secretion of IL-12p40 by PAO was reduced by forskolin, a cAMP activator. These results suggest that PAO enhances IL-12p40 production by inhibiting CRE activity.
Collapse
|
31
|
Kundu S, Fan K, Cao M, Lindner DJ, Zhao ZJ, Borden E, Yi T. Novel SHP-1 inhibitors tyrosine phosphatase inhibitor-1 and analogs with preclinical anti-tumor activities as tolerated oral agents. THE JOURNAL OF IMMUNOLOGY 2010; 184:6529-36. [PMID: 20421638 DOI: 10.4049/jimmunol.0903562] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Src homology region 2 domain-containing phosphatase 1 (SHP-1) has been implicated as a potential cancer therapeutic target by its negative regulation of immune cell activation and the activity of the SHP-1 inhibitor sodium stibogluconate that induced IFN-gamma(+) cells for anti-tumor action. To develop more potent SHP-1-targeted anti-cancer agents, inhibitory leads were identified from a library of 34,000 drug-like compounds. Among the leads and active at low nM for recombinant SHP-1, tyrosine phosphatase inhibitor-1 (TPI-1) selectively increased SHP-1 phospho-substrates (pLck-pY394, pZap70, and pSlp76) in Jurkat T cells but had little effects on pERK1/2 or pLck-pY505 regulated by phosphatases SHP-2 or CD45, respectively. TPI-1 induced mouse splenic-IFN-gamma(+) cells in vitro, approximately 58-fold more effective than sodium stibogluconate, and increased mouse splenic-pLck-pY394 and -IFN-gamma(+) cells in vivo. TPI-1 also induced IFN-gamma(+) cells in human peripheral blood in vitro. Significantly, TPI-1 inhibited ( approximately 83%, p < 0.002) the growth of B16 melanoma tumors in mice at a tolerated oral dose in a T cell-dependent manner but had little effects on B16 cell growth in culture. TPI-1 also inhibited B16 tumor growth and prolonged tumor mice survival as a tolerated s.c. agent. TPI-1 analogs were identified with improved activities in IFN-gamma(+) cell induction and in anti-tumor actions. In particular, analog TPI-1a4 as a tolerated oral agent completely inhibited the growth of K1735 melanoma tumors and was more effective than the parental lead against MC-26 colon cancer tumors in mice. These results designate TPI-1 and the analogs as novel SHP-1 inhibitors with anti-tumor activity likely via an immune mechanism, supporting SHP-1 as a novel target for cancer treatment.
Collapse
Affiliation(s)
- Suman Kundu
- Department of Cancer Biology, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Ahmed Z, George R, Lin CC, Suen KM, Levitt JA, Suhling K, Ladbury JE. Direct binding of Grb2 SH3 domain to FGFR2 regulates SHP2 function. Cell Signal 2010; 22:23-33. [DOI: 10.1016/j.cellsig.2009.08.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 08/21/2009] [Accepted: 08/27/2009] [Indexed: 01/08/2023]
|
33
|
Zhou X, Agazie YM. Molecular mechanism for SHP2 in promoting HER2-induced signaling and transformation. J Biol Chem 2009; 284:12226-34. [PMID: 19261604 DOI: 10.1074/jbc.m900020200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Src homology phosphotyrosyl phosphatase 2 (SHP2) plays a positive role in HER2-induced signaling and transformation, but its mechanism of action is poorly understood. Given the significance of HER2 in breast cancer, defining a mechanism for SHP2 in the HER2 signaling pathway is of paramount importance. In the current report we show that SHP2 positively modulates the Ras-extracellular signal-regulated kinase 1 and 2 and the phospoinositide-3-kinase-Akt pathways downstream of HER2 by increasing the half-life the activated form of Ras. This is accomplished by dephosphorylating an autophosphorylation site on HER2 that serves as a docking platform for the SH2 domains of the Ras GTPase-activating protein (RasGAP). The net effect is an increase in the intensity and duration of GTP-Ras levels with the overall impact of enhanced HER2 signaling and cell transformation. In conformity to these findings, the HER2 mutant that lacks the SHP2 target site exhibits an enhanced signaling and cell transformation potential. Therefore, SHP2 promotes HER2-induced signaling and transformation at least in part by dephosphorylating a negative regulatory autophosphorylation site. These results suggest that SHP2 might serve as a therapeutic target against breast cancer and other cancers characterized by HER2 overexpression.
Collapse
Affiliation(s)
- Xiangdong Zhou
- Department of Biochemistry and The Marry Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia 26506, USA
| | | |
Collapse
|
34
|
Abstract
Tyrosine phosphorylation and dephosphorylation of proteins play a critical role for many T-cell functions. The opposing actions of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) determine the level of tyrosine phosphorylation at any time. It is well accepted that PTKs are essential during T-cell signaling; however, the role and importance of PTPs are much less known and appreciated. Both transmembrane and cytoplasmic tyrosine phosphatases have been identified in T cells and shown to regulate T-cell responses. This review focuses on the roles of the two cytoplasmic PTPs, the Src-homology 2 domain (SH2)-containing SHP-1 and SHP-2, in T-cell signaling, development, differentiation, and function.
Collapse
Affiliation(s)
- Ulrike Lorenz
- Department of Microbiology and The Beirne Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908-0734, USA.
| |
Collapse
|
35
|
Young JA, Becker AM, Medeiros JJ, Shapiro VS, Wang A, Farrar JD, Quill TA, van Huijsduijnen RH, van Oers NS. The protein tyrosine phosphatase PTPN4/PTP-MEG1, an enzyme capable of dephosphorylating the TCR ITAMs and regulating NF-kappaB, is dispensable for T cell development and/or T cell effector functions. Mol Immunol 2008; 45:3756-66. [PMID: 18614237 PMCID: PMC2596642 DOI: 10.1016/j.molimm.2008.05.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 05/27/2008] [Accepted: 05/28/2008] [Indexed: 11/16/2022]
Abstract
T cell receptor signaling processes are controlled by the integrated actions of families of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPases). Several distinct cytosolic protein tyrosine phosphatases have been described that are able to negatively regulate TCR signaling pathways, including SHP-1, SHP-2, PTPH1, and PEP. Using PTPase substrate-trapping mutants and wild type enzymes, we determined that PTPN4/PTP-MEG1, a PTPH1-family member, could complex and dephosphorylate the ITAMs of the TCR zeta subunit. In addition, the substrate-trapping derivative augmented basal and TCR-induced activation of NF-kappaB in T cells. To characterize the contribution of this PTPase in T cells, we developed PTPN4-deficient mice. T cell development and TCR signaling events were comparable between wild type and PTPN4-deficient animals. The magnitude and duration of TCR-regulated ITAM phosphorylation, as well as overall protein phosphorylation, was unaltered in the absence of PTPN4. Finally, Th1- and Th2-derived cytokines and in vivo immune responses to Listeria monocytogenes were equivalent between wild type and PTPN4-deficient mice. These findings suggest that additional PTPases are involved in controlling ITAM phosphorylations.
Collapse
Affiliation(s)
- Jennifer A. Young
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Amy M. Becker
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jennifer J. Medeiros
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Virginia S. Shapiro
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Andrew Wang
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - J. David Farrar
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Timothy A. Quill
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Nicolai S.C. van Oers
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
36
|
Zhou X, Coad J, Ducatman B, Agazie YM. SHP2 is up-regulated in breast cancer cells and in infiltrating ductal carcinoma of the breast, implying its involvement in breast oncogenesis. Histopathology 2008; 53:389-402. [PMID: 18643929 DOI: 10.1111/j.1365-2559.2008.03103.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS To determine whether Src homology phosphotyrosyl phosphatase 2 (SHP2) is up-regulated in breast cancer and, if so, to determine whether its up-regulation has any relationship with clinical variables of breast cancer. METHODS AND RESULTS Immunoblotting, immunohistochemistry and immunofluorescence microscopy were used to assess the state of SHP2 expression in breast cancer cells and in infiltrating ductal carcinoma (IDC) of breast. The possible role of SHP2 in breast cancer cell transformation was determined by dominant-negative expression and anchorage-independent growth assays. All of the breast cancer cell lines tested and 72% of IDC breast tumours analysed had increased amounts of the SHP2 protein. In support of its positive role, dominant-negative SHP2 blocked anchorage-independent growth of breast cancer cells. Furthermore, overexpression of SHP2 seemed to have a positive relationship to HER2 overexpression, nuclear accumulation of hormone receptors, higher tumour grade and lymph node metastasis, but not to age of breast cancer patients. CONCLUSION SHP2 is a widely overexpressed signalling protein in IDC breast tumours. Given SHP2's positive role in cell growth, transformation and stem cell survival, the positive relationship of its overexpression to lymph node metastasis, nuclear accumulation of hormone receptors and higher tumour grade suggests that SHP2 promotes breast oncogenesis.
Collapse
Affiliation(s)
- X Zhou
- Department of Biochemistry and Molecular Biology, West Virginia University, Morgantown, WV 26506, USA
| | | | | | | |
Collapse
|
37
|
Lee HSW, Ostrowski MA, Gray-Owen SD. CEACAM1 dynamics during neisseria gonorrhoeae suppression of CD4+ T lymphocyte activation. THE JOURNAL OF IMMUNOLOGY 2008; 180:6827-35. [PMID: 18453603 DOI: 10.4049/jimmunol.180.10.6827] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neisseria gonorrhoeae colony opacity-associated (Opa) proteins bind to human carcinoembryonic antigen cellular adhesion molecules (CEACAM) found on host cells including T lymphocytes. Opa binding to CEACAM1 suppresses the activation of CD4(+) T cells in response to a variety of stimuli. In this study, we use primary human CD4(+) T cells isolated from peripheral blood to define the molecular events occurring subsequent to Opa-CEACAM1 binding. We establish that, in contrast to other cell types, T cells do not engulf N. gonorrhoeae upon CEACAM1 binding. Instead, the bacteria recruit CEACAM1 from intracellular stores and maintain it on the T cell surface. Upon TCR ligation, the co-engaged CEACAM1 becomes phosphorylated on tyrosine residues within the ITIMs apparent in the cytoplasmic domain. This allows the recruitment and subsequent activation of the src homology domain 2-containing tyrosine phosphatases SHP-1 and SHP-2 at the site of bacterial attachment, which prevents the normal tyrosine phosphorylation of the CD3zeta-chain and ZAP-70 kinase in response to TCR engagement. Combined, this dynamic response allows the bacteria to effectively harness the coinhibitory function of CEACAM1 to suppress the adaptive immune response at its earliest step.
Collapse
Affiliation(s)
- Hannah S W Lee
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
38
|
Zhou XD, Agazie YM. Inhibition of SHP2 leads to mesenchymal to epithelial transition in breast cancer cells. Cell Death Differ 2008; 15:988-96. [DOI: 10.1038/cdd.2008.54] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
39
|
Phosphatase SHP-1 promotes TLR- and RIG-I-activated production of type I interferon by inhibiting the kinase IRAK1. Nat Immunol 2008; 9:542-50. [PMID: 18391954 DOI: 10.1038/ni.1604] [Citation(s) in RCA: 212] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Accepted: 03/11/2008] [Indexed: 11/08/2022]
Abstract
Unbalanced production of proinflammatory cytokines and type I interferons in immune responses may lead to immunopathology; thus, the mechanisms that ensure the beneficial production of proinflammatory cytokines and type I interferons are of particular importance. Here we demonstrate that the phosphatase SHP-1 negatively regulated Toll-like receptor-mediated production of proinflammatory cytokines by inhibiting activation of the transcription factor NF-kappaB and mitogen-activated protein kinase. Simultaneously, SHP-1 increased the production of type I interferon mediated by Toll-like receptors and the helicase RIG-I by directly binding to and inhibiting activation of the kinase IRAK1. Our data demonstrate that SHP-1 contributes to immune homeostasis by balancing the production of proinflammatory cytokines and type I interferons in the innate immune response.
Collapse
|
40
|
Abstract
Activation of the T-cell co-receptor cytotoxic T-lymphocyte antigen 4 (CTLA4) has a pivotal role in adjusting the threshold for T-cell activation and in preventing autoimmunity and massive tissue infiltration by T cells. Although many mechanistic models have been postulated, no single model has yet accounted for its overall function. In this Opinion article, I outline the strengths and weaknesses of the current models, and present a new 'reverse stop-signal model' to account for CTLA4 function.
Collapse
Affiliation(s)
- Christopher E Rudd
- Cell Signalling Section, Division of Immunology, Department of Pathology, Tennis Court Road, CB1 4QP Cambridge, UK.
| |
Collapse
|
41
|
Mitsuhashi M, Ogura M, Endo K, Obara K, Izutsu H, Targan SR, Maemura M, Tachikawa D, Shinagawa A. Ex vivo induction of mRNA in human whole blood as a new platform of drug and dietary supplement development. Pharm Res 2008; 25:1116-24. [PMID: 18183479 PMCID: PMC2292497 DOI: 10.1007/s11095-007-9510-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Accepted: 11/21/2007] [Indexed: 11/29/2022]
Abstract
Purpose We introduced a new concept of ex vivo gene expression analysis (Mitsuhashi, Clin Chem 53:148–149, 2007), where drug action was simulated under physiological conditions. This model system was applied to study various fields of drug development. Materials and Methods Heparinized human whole blood was incubated with drugs for less than 4h. The changes of specific mRNA were then quantified using the method we developed (Mitsuhashi, Tomozawa, Endo, and Shinagawa, Clin Chem 52:634–642, 2006). Results The mRNA quantitation method was used as a model system to study the following areas: (1) identification of respondents and non-respondents, (2) ex vivo compound screening, (3) determination of individually optimized doses, (4) drug-to-drug comparison, (5) assessment of leukocyte toxicity, (6) discovery of molecular targets, (7) assessment of the action of dietary supplements, and (8) characterization of respondents and non-respondents for various dietary supplements. Conclusion Since ex vivo assays are safe, a large number of healthy donors and disease patients can be recruited to identify individual-to-individual variations, which is not available from current preclinical study models. Although each system should be validated using a large number of samples, the ex vivo analysis will be a new tool for the development of drugs and dietary supplements in future. Electronic supplementary material The online version of this article (doi:10.1007/s11095-007-9510-2) contains supplementary material, which is available to authorized users.
Collapse
|
42
|
SRC family kinases and receptors: analysis of three activation mechanisms by dynamic systems modeling. Biophys J 2007; 94:1995-2006. [PMID: 18055537 DOI: 10.1529/biophysj.107.115022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Src family kinases (SFKs) interact with a number of cellular receptors. They participate in diverse signaling pathways and cellular functions. Most of the receptors involved in SFK signaling are characterized by similar modes of regulation. This computational study discusses a general kinetic model of SFK-receptor interaction. The analysis of the model reveals three major ways of SFK activation: release of inhibition by C-terminal Src kinase, weakening of the inhibitory intramolecular phosphotyrosine-SH2 interaction, and amplification of a stimulating kinase activity. The SFK model was then extended to simulate interaction with growth factor and T-cell receptors. The modular SFK signaling system was shown to adapt to the requirements of specific signaling contexts and yield qualitatively different responses in the different simulated environments. The model also provides a systematic overview of the major interactions between SFKs and various cellular signaling systems and identifies their common properties.
Collapse
|
43
|
Ma CS, Nichols KE, Tangye SG. Regulation of cellular and humoral immune responses by the SLAM and SAP families of molecules. Annu Rev Immunol 2007; 25:337-79. [PMID: 17201683 DOI: 10.1146/annurev.immunol.25.022106.141651] [Citation(s) in RCA: 206] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
SAP (SLAM-associated protein) was identified in 1998 as an adaptor molecule involved in the intracellular signaling pathways elicited through the cell surface receptor SLAM and as the protein defective in the human immunodeficiency X-linked lymphoproliferative disease (XLP). During the past eight years, it has been established that the SLAM family of cell surface receptors (SLAM, 2B4, NTB-A, Ly9, CD84) and the SAP family of adaptors (SAP, EAT-2, ERT) play critical roles in lymphocyte development, differentiation, and acquisition of effector functions. Studies of these proteins have shown unexpected roles in cytokine production by T cells and myeloid cells, T cell-dependent humoral immune responses, NK cell-mediated cytotoxicity, and NKT cell development. This review highlights recent findings that have improved our understanding of the roles of the SLAM and SAP families of molecules in immune regulation and discusses how perturbations in the signaling pathways involving these proteins can result in different disease states.
Collapse
Affiliation(s)
- Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, 2010, New South Wales, Australia.
| | | | | |
Collapse
|
44
|
Hoff H, Brunner-Weinzierl MC. The tyrosine phosphatase SHP-2 regulates differentiation and apoptosis of individual primary T lymphocytes. Eur J Immunol 2007; 37:1072-86. [PMID: 17330819 DOI: 10.1002/eji.200636240] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although phosphatases are key players of intracellular processes, not much is known about the phosphatase SHP-2 during T cell differentiation. Here we show that ectopic over-expression of SHP-2 in primary T helper cells directly reduced the frequency of individual lymphocytes expressing pro-inflammatory cytokines after antigen-specific stimulation by a mechanism impairing activation of protein kinase C. In addition we demonstrate that SHP-2 mediates enhanced migration upon CXCR4 signaling in a G-protein-dependent manner. Most strikingly, SHP-2 mediated a dramatic increase in apoptosis by highly enhanced activation of caspases. Co-immunoprecipitations of SHP-2 and c-Cbl from primary T helper cells demonstrated that SHP-2 strongly interacts with the ubiquitin ligase c-Cbl, indicating that c-Cbl could mediate the negative signals of SHP-2. Our results show that SHP-2 signal transduction regulates central checkpoints of T cell differentiation by the activation of distinct signaling cascades.
Collapse
Affiliation(s)
- Holger Hoff
- Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | | |
Collapse
|
45
|
Abstract
Ras genes are the most common targets for somatic gain-of-function mutations in human cancer. Recently, germline mutations that affect components of the Ras-Raf-mitogen-activated and extracellular-signal regulated kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway were shown to cause several developmental disorders, including Noonan, Costello and cardio-facio-cutaneous syndromes. Many of these mutant alleles encode proteins with aberrant biochemical and functional properties. Here we will discuss the implications of germline mutations in the Ras-Raf-MEK-ERK pathway for understanding normal developmental processes and cancer pathogenesis.
Collapse
Affiliation(s)
- Suzanne Schubbert
- Department of Pediatrics, University of California, 513 Parnassus Avenue, Room HSE-302, San Francisco, California 94143, USA
| | | | | |
Collapse
|
46
|
Abstract
Tyrosyl phosphorylation plays a critical role in multiple signaling pathways regulating innate and acquired immunity. Although tyrosyl phosphorylation is a reversible process, we know much more about the functions of protein-tyrosine kinases (PTKs) than about protein-tyrosine phosphatases (PTPs). Genome sequencing efforts have revealed a large and diverse superfamily of PTPs, which can be subdivided into receptor-like (RPTPs) and nonreceptor (NRPTPs). The role of the RPTP CD45 in immune cell signaling is well known, but those of most other PTPs remain poorly understood. Here, we review the mechanism of action, regulation, and physiological functions of NRPTPs in immune cell signaling. Such an analysis indicates that PTPs are as important as PTKs in regulating the immune system.
Collapse
Affiliation(s)
- Lily I Pao
- Cancer Biology Program, Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA.
| | | | | | | |
Collapse
|
47
|
Dar WA, Knechtle SJ. CXCR3-mediated T-cell chemotaxis involves ZAP-70 and is regulated by signalling through the T-cell receptor. Immunology 2007; 120:467-85. [PMID: 17250586 PMCID: PMC2265907 DOI: 10.1111/j.1365-2567.2006.02534.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The chemokine receptor CXCR3 is critical for the function of activated T cells. We studied the molecular mechanisms of CXCR3 signalling. The addition of CXCR3 ligands to normal human T cells expressing CXCR3 led to the tyrosine phosphorylation of multiple proteins. Addition of the same ligands to Jurkat T cells engineered to express CXCR3 induced tyrosine phosphorylation of proteins with molecular weights similar to those in normal cells. Immunoblotting with phosphotyrosine-specific antibodies identified Zeta-associated protein of 70,000 molecular weight (ZAP-70), linker for the activation of T cells (LAT), and phospholipase-C-gamma1 (PLCgamma1) to be among the proteins that become phosphorylated upon CXCR3 activation. ZAP-70 was phosphorylated on tyrosine 319, LAT on tyrosines 171 and 191, and PLCgamma1 on tyrosine 783. The ZAP-70 inhibitor piceatannol reduced CXCR3-mediated tyrosine phosphorylation of ZAP-70, LAT, PLCgamma1 and mitogen-activated protein kinase Erk and it reduced CXCL10-mediated chemotaxis of both CXCR3-transfected Jurkat T cells and normal T cells expressing CXCR3. These results are consistent with the involvement of ZAP-70 in CXCR3-mediated protein tyrosine phosphorylation and CXCR3-induced T-cell chemotaxis. Studies with the Lck-deficient Jurkat T-cell line, JCAM1.6, demonstrated that phosphorylation of ZAP-70 after CXCR3 activation is a Lck-dependent process. Finally, stimulating CXCR3-expressing Jurkat T cells and normal T cells expressing CXCR3 through the T-cell receptor attenuated CXCR3-induced tyrosine phosphorylation and CXCR3-mediated T-cell migration, indicating the occurrence of cross-talk between T-cell receptor and CXCR3-signalling pathways. These results shed light on the mechanisms of CXCR3 signalling. Such information could be useful when designing therapeutic strategies to regulate T-cell function.
Collapse
Affiliation(s)
- Wasim A Dar
- Department of Surgery, Division of Transplantation, University of Wisconsin-Madison, Madison, WI 53792-7375, USA.
| | | |
Collapse
|
48
|
Saunders PA, Hendrycks VR, Lidinsky WA, Woods ML. PD-L2:PD-1 involvement in T cell proliferation, cytokine production, and integrin-mediated adhesion. Eur J Immunol 2006; 35:3561-9. [PMID: 16278812 DOI: 10.1002/eji.200526347] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The B7 family member programmed death ligand 2 (PD-L2) has been implicated in both positive and negative regulation of T cell activity. In this study, we demonstrate that on human T cells, PD-L2 acts only as a negative regulator of T cell activity, inhibiting proliferation, IL-2 production, and IFN-gamma production via its interaction with programmed death-1 (PD-1). This study also shows a novel role for PD-1 in inhibiting beta1 and beta2 integrin-mediated adhesion. PD-L2 inhibition of T cell function involves modulation of the phosphoinositide 3-OH kinase (PI 3-K)/AKT and extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) pathways, with PD-L2 inhibiting anti-CD3-induced AKT phosphorylation within minutes and ERK phosphorylation after hours. Analysis of phosphatase activity of Src homology 2 domain-containing tyrosine phosphatase (SHP)-1 and SHP-2 in response to anti-CD3 mAb or anti-CD3 mAb + PD-L2 stimulation revealed that while SHP-1 phosphatase activity is not affected by stimulation, SHP-2 phosphatase activity is significantly increased by anti-CD3 mAb + PD-L2 stimulation. Anti-CD3 mAb + PD-L2 stimulation also increased the level of SHP-2 associated with the PD-1 receptor. These results suggest that catalytically active SHP-2 associated with the PD-1 receptor is involved in modulating T cell function.
Collapse
Affiliation(s)
- Paul A Saunders
- Bioassay Department, R&D Systems, Inc., Minneapolis, MN 55413, USA
| | | | | | | |
Collapse
|
49
|
Abstract
Within the paradigm of the two-signal model of lymphocyte activation, the interest in costimulation has witnessed a remarkable emergence in the past few years with the discovery of a large array of molecules that can serve this role, including some with an inhibitory function. Interest has been further enhanced by the realization of these molecules' potential as targets to modulate clinical immune responses. Although the therapeutic translation of mechanistic knowledge in costimulatory molecules has been relatively straightforward, the capacity to target their inhibitory counterparts has remained limited. This limited capacity is particularly apparent in the case of the cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), a major negative regulator of T cell responses. Because there have been several previous comprehensive reviews on the function of this molecule, we focus here on the physiological implications of its structural features. Such an exercise may ultimately help us to design immunotherapeutic agents that target CTLA-4.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, CD
- Antigens, Differentiation/chemistry
- Antigens, Differentiation/genetics
- Antigens, Differentiation/metabolism
- Biological Transport, Active
- CTLA-4 Antigen
- Dimerization
- Evolution, Molecular
- Humans
- Ligands
- Lymphocyte Activation
- Models, Immunological
- Molecular Biology
- Molecular Sequence Data
- Polymorphism, Genetic
- Protein Structure, Quaternary
- Sequence Homology, Amino Acid
- Signal Transduction
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Wendy A Teft
- The FOCIS Center for Clinical Immunology and Immunotherapeutics, Robarts Research Institute, and Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada, N6A 5K8
| | | | | |
Collapse
|
50
|
Burks J, Agazie YM. Modulation of alpha-catenin Tyr phosphorylation by SHP2 positively effects cell transformation induced by the constitutively active FGFR3. Oncogene 2006; 25:7166-79. [PMID: 16767162 DOI: 10.1038/sj.onc.1209728] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The Src homology 2 phosphotyrosyl phosphatase (SHP2) is a nonreceptor-type phosphatase that acts as a positive transducer of receptor Tyr kinase (RTK) signaling, particularly the Ras-REK and PI3K-Akt pathways. Recently, we have demonstrated that SHP2 is required for cell transformation induced by the constitutively active fibroblast growth factor receptor 3 (K/E-FR3) (Oncogene, 22, 6909-6918). In that study, we had detected a phosphotyrosyl protein of approximately 100 KDa (p100) in cells expressing dominant-negative SHP2 (R/E-SHP2), but its identity and relevance in SHP2-meditaed transformation was not known. Here, we report the identification of p100 as alpha-catenin, a vinculin-related protein involved in adherens junction-mediated intercellular adhesion. We show that alpha-catenin becomes Tyr phosphorylated in intercellular adhesion-dependent manner and this event is counteracted by SHP2. Substrate trapping in intact cells and immunocomplex phosphatse assays confirmed that alpha-catenin is in deed an SHP2 substrate. Tyr phosphorylation of alpha-catenin enhances its translocation to the plasma membrane and its interaction with beta-catenin, leading to enhanced actin polymerization and stabilization of adherens junction-mediated intercellular adhesion, a phenomenon commensurate with loss of the transformation phenotype. Site-directed mutagenesis studies also suggested that Tyr phosphorylation of alpha-catenin enhances its inhibitory role on cell transformation. Based on our previous work and the current report, we demonstrate that mediation of cell transformation by SHP2 is a complex process that involves modulation of the Ras-ERK and PI3K-Akt signaling pathways, intercellular adhesion, focal adhesion and actin cytoskeletal reorganization. To our knowledge, this is the first report showing regulation of alpha-catenin function by Tyr phosphorylation and its inhibitory effect on cell transformation.
Collapse
Affiliation(s)
- J Burks
- Department of Biochemistry and Molecular Pharmacology, Robert C Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506-9142, USA
| | | |
Collapse
|