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
|
Wang Q, Li H, Mao Y, Garg A, Park ES, Wu Y, Chow A, Peregrin J, Zhang X. Shc1 cooperates with Frs2 and Shp2 to recruit Grb2 in FGF-induced lens development. eLife 2025; 13:RP103615. [PMID: 40327534 PMCID: PMC12055001 DOI: 10.7554/elife.103615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
Fibroblast growth factor (FGF) signaling elicits multiple downstream pathways, most notably the Ras/MAPK cascade facilitated by the adaptor protein Grb2. However, the mechanism by which Grb2 is recruited to the FGF signaling complex remains unresolved. Here, we showed that genetic ablation of FGF signaling prevented murine lens induction by disrupting transcriptional regulation and actin cytoskeletal arrangements, which could be reproduced by deleting the juxtamembrane region of the FGF receptor and rescued by Kras activation. Conversely, mutations affecting the Frs2-binding site on the FGF receptor or the deletion of Frs2 and Shp2 primarily impact later stages of lens vesicle development involving lens fiber cell differentiation. Our study further revealed that the loss of Grb2 abolished MAPK signaling, resulting in a profound arrest of lens development. However, removing Grb2's putative Shp2 dephosphorylation site (Y209) neither produced a detectable phenotype nor impaired MAPK signaling during lens development. Furthermore, the catalytically inactive Shp2 mutation (C459S) only modestly impaired FGF signaling, whereas replacing Shp2's C-terminal phosphorylation sites (Y542/Y580) previously implicated in Grb2 binding only caused placental defects, perinatal lethality, and reduced lacrimal gland branching without impacting lens development, suggesting that Shp2 only partially mediates Grb2 recruitment. In contrast, we observed that FGF signaling is required for the phosphorylation of the Grb2-binding sites on Shc1 and the deletion of Shc1 exacerbates the lens vesicle defect caused by Frs2 and Shp2 deletion. These findings establish Shc1 as a critical collaborator with Frs2 and Shp2 in targeting Grb2 during FGF signaling.
Collapse
Affiliation(s)
- Qian Wang
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
| | - Hongge Li
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
| | - Yingyu Mao
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
| | - Ankur Garg
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
| | - Eun Sil Park
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
| | - Yihua Wu
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
| | - Alyssa Chow
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
| | - John Peregrin
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
| | - Xin Zhang
- Department of Ophthalmology, Columbia UniversityNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia UniversityNew YorkUnited States
| |
Collapse
|
3
|
Wang Q, Li H, Mao Y, Garg A, Park ES, Wu Y, Chow A, Peregrin J, Zhang X. Shc1 cooperates with Frs2 and Shp2 to recruit Grb2 in FGF-induced lens development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.20.619055. [PMID: 39484547 PMCID: PMC11527007 DOI: 10.1101/2024.10.20.619055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Fibroblast growth factor (FGF) signaling elicits multiple downstream pathways, most notably the Ras/MAPK cascade facilitated by the adaptor protein Grb2. However, the mechanism by which Grb2 is recruited to the FGF signaling complex remains unresolved. Here we showed that genetic ablation of FGF signaling prevented lens induction by disrupting transcriptional regulation and actin cytoskeletal arrangements, which could be reproduced by deleting the juxtamembrane region of the FGF receptor and rescued by Kras activation. Conversely, mutations affecting the Frs2-binding site on the FGF receptor or the deletion of Frs2 and Shp2 primarily impact later stages of lens vesicle development involving lens fiber cell differentiation. Our study further revealed that the loss of Grb2 abolished MAPK signaling, resulting in a profound arrest of lens development. However, removing Grb2's putative Shp2 dephosphorylation site (Y209) neither produced a detectable phenotype nor impaired MAPK signaling during lens development. Furthermore, the catalytically inactive Shp2 mutation (C459S) only modestly impaired FGF signaling, whereas replacing Shp2's C-terminal phosphorylation sites (Y542/Y580) previously implicated in Grb2 binding only caused placental defects, perinatal lethality, and reduced lacrimal gland branching without impacting lens development, suggesting that Shp2 only partially mediates Grb2 recruitment. In contrast, we observed that FGF signaling is required for the phosphorylation of the Grb2-binding sites on Shc1 and the deletion of Shc1 exacerbates the lens vesicle defect caused by Frs2 and Shp2 deletion. These findings establish Shc1 as a critical collaborator with Frs2 and Shp2 in targeting Grb2 during FGF signaling.
Collapse
Affiliation(s)
- Qian Wang
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Hongge Li
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Yingyu Mao
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Ankur Garg
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Eun Sil Park
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Yihua Wu
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Alyssa Chow
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - John Peregrin
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Xin Zhang
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|
4
|
Van Dyck PK, Piszkin L, Gorski EA, Nascimento ET, Abebe JA, Hoffmann LM, Peng JW, White KA. Ionizable networks mediate pH-dependent allostery in SH2 signaling proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608875. [PMID: 39229188 PMCID: PMC11370553 DOI: 10.1101/2024.08.21.608875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
IntroductionTransient intracellular pH dynamics1regulate mammalian proliferation2,3, migration4, and differentiation5. However, for many pH-dependent cell processes, the molecular mediators are unknown6. Prior work identified histidine residues as molecular switches in pH-sensitive proteins, but how other ionizable residues contribute to pH-dependent protein allostery is understudied. Here, we develop anin silicocomputational pipeline to identify putative pH-sensitive proteins and their molecular mechanisms. We first apply this pipeline to SHP2, a known pH-sensitive signaling protein with an uncharacterized molecular mechanism. We show wild-type SHP2 phosphatase activity is pH-sensitivein vitroand in cells, and mutation of identified H116 and E252 to non-titratable alanine residues abolishes pH-sensitive function. We also show that c-Src is a previously unrecognized pH-dependent kinase, and mutation of the identified ionizable network again abolishes pH-sensitive activity. Constant pH molecular dynamics simulations support a conserved allosteric mechanism of pH-dependent binding of inhibitory SH2 domains to the functional catalytic domains of SHP2 and c-Src. We apply our computational pipeline across SH2 domain-containing signaling proteins and identify evolutionarily conserved putative pH-sensing networks. Our results reveal that pH is an allosteric regulator of SH2 domain-containing signaling proteins providing insight into normal pH-dependent cell biology and diseases where pHi is dysregulated, such as cancer.
Collapse
Affiliation(s)
- Papa Kobina Van Dyck
- Department of Chemistry and Biochemistry, University of Notre Dame 251 Nieuwland Science Hall Notre Dame, IN 46556 USA
- Harper Cancer Research Institute, University of Notre Dame 1234 N. Notre Dame Avenue South Bend, IN 46617 USA
| | - Luke Piszkin
- Department of Chemistry and Biochemistry, University of Notre Dame 251 Nieuwland Science Hall Notre Dame, IN 46556 USA
| | - Elijah A. Gorski
- Department of Chemistry and Biochemistry, University of Notre Dame 251 Nieuwland Science Hall Notre Dame, IN 46556 USA
- Harper Cancer Research Institute, University of Notre Dame 1234 N. Notre Dame Avenue South Bend, IN 46617 USA
| | - Eduarda Tartarella Nascimento
- Department of Chemistry and Biochemistry, University of Notre Dame 251 Nieuwland Science Hall Notre Dame, IN 46556 USA
- Harper Cancer Research Institute, University of Notre Dame 1234 N. Notre Dame Avenue South Bend, IN 46617 USA
- Saint Mary’s College, Notre Dame, IN 46556 USA
| | - Joshua A. Abebe
- Department of Chemistry and Biochemistry, University of Notre Dame 251 Nieuwland Science Hall Notre Dame, IN 46556 USA
- Harper Cancer Research Institute, University of Notre Dame 1234 N. Notre Dame Avenue South Bend, IN 46617 USA
| | - Logan M. Hoffmann
- Department of Chemistry and Biochemistry, University of Notre Dame 251 Nieuwland Science Hall Notre Dame, IN 46556 USA
- Harper Cancer Research Institute, University of Notre Dame 1234 N. Notre Dame Avenue South Bend, IN 46617 USA
| | - Jeffrey W. Peng
- Department of Chemistry and Biochemistry, University of Notre Dame 251 Nieuwland Science Hall Notre Dame, IN 46556 USA
| | - Katharine A. White
- Department of Chemistry and Biochemistry, University of Notre Dame 251 Nieuwland Science Hall Notre Dame, IN 46556 USA
- Harper Cancer Research Institute, University of Notre Dame 1234 N. Notre Dame Avenue South Bend, IN 46617 USA
| |
Collapse
|
5
|
de Jesus VHF, Mathias-Machado MC, de Farias JPF, Aruquipa MPS, Jácome AA, Peixoto RD. Targeting KRAS in Pancreatic Ductal Adenocarcinoma: The Long Road to Cure. Cancers (Basel) 2023; 15:5015. [PMID: 37894382 PMCID: PMC10605759 DOI: 10.3390/cancers15205015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains an important cause of cancer-related mortality, and it is expected to play an even bigger part in cancer burden in the years to come. Despite concerted efforts from scientists and physicians, patients have experienced little improvement in survival over the past decades, possibly because of the non-specific nature of the tested treatment modalities. Recently, the discovery of potentially targetable molecular alterations has paved the way for the personalized treatment of PDAC. Indeed, the central piece in the molecular framework of PDAC is starting to be unveiled. KRAS mutations are seen in 90% of PDACs, and multiple studies have demonstrated their pivotal role in pancreatic carcinogenesis. Recent investigations have shed light on the differences in prognosis as well as therapeutic implications of the different KRAS mutations and disentangled the relationship between KRAS and effectors of downstream and parallel signaling pathways. Additionally, the recognition of other mechanisms involving KRAS-mediated pathogenesis, such as KRAS dosing and allelic imbalance, has contributed to broadening the current knowledge regarding this molecular alteration. Finally, KRAS G12C inhibitors have been recently tested in patients with pancreatic cancer with relative success, and inhibitors of KRAS harboring other mutations are under clinical development. These drugs currently represent a true hope for a meaningful leap forward in this dreadful disease.
Collapse
Affiliation(s)
| | | | | | | | - Alexandre A. Jácome
- Department of Gastrointestinal Medical Oncology, Oncoclínicas, Belo Horizonte 30360-680, Brazil
| | | |
Collapse
|
6
|
Li Y, Zhou H, Liu P, Lv D, Shi Y, Tang B, Xu J, Zhong T, Xu W, Zhang J, Zhou J, Ying K, Zhao Y, Sun Y, Jiang Z, Cheng H, Zhang X, Ke Y. SHP2 deneddylation mediates tumor immunosuppression in colon cancer via the CD47/SIRPα axis. J Clin Invest 2023; 133:162870. [PMID: 36626230 PMCID: PMC9927946 DOI: 10.1172/jci162870] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
SIPRα on macrophages binds with CD47 to resist proengulfment signals, but how the downstream signal of SIPRα controls tumor-infiltrating macrophages (TIMs) is still poorly clarified. Here, we report that the CD47/signal regulatory protein α (SIRPα) axis requires the deneddylation of tyrosine phosphatase SHP2. Mechanistically, Src homology region 2-containing protein tyrosine phosphatase 2 (SHP2) was constitutively neddylated on K358 and K364 sites; thus, its autoinhibited conformation was maintained. In response to CD47-liganded SIRPα, SHP2 was deneddylated by sentrin-specific protease 8 (SENP8), which led to the dephosphorylation of relevant substrates at the phagocytic cup and subsequent inhibition of macrophage phagocytosis. Furthermore, neddylation inactivated myeloid-SHP2 and greatly boosted the efficacy of colorectal cancer (CRC) immunotherapy. Importantly, we observed that supplementation with SHP2 allosteric inhibitors sensitized immune treatment-resistant CRC to immunotherapy. Our results emphasize that the CRC subtype that is unresponsive to immunotherapy relies on SIRPαhiSHP2hiNEDD8lo TIMs and highlight the need to further explore the strategy of SHP2 targeting in CRC therapy.
Collapse
Affiliation(s)
- Yiqing Li
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Zhou
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Liu
- Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Dandan Lv
- Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yichun Shi
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bufu Tang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research at The Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Jiaqi Xu
- Department of Pathology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangou, China
| | - Tingting Zhong
- Department of Pathology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangou, China
| | - Wangting Xu
- Department of Respiratory Medicine at The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Zhang
- Department of Urology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Medicine at The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kejing Ying
- Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchao Zhao
- Cancer Institute of The Second Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Sun
- Cancer Institute of The Second Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhinong Jiang
- Department of Pathology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangou, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, and Department of Cardiology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuehai Ke
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 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
|
Ryan A, Janosko CP, Courtney TM, Deiters A. Engineering SHP2 Phosphatase for Optical Control. Biochemistry 2022; 61:2687-2697. [DOI: 10.1021/acs.biochem.2c00387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Amy Ryan
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Chasity P. Janosko
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Taylor M. Courtney
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
9
|
Solman M, Woutersen DTJ, den Hertog J. Modeling (not so) rare developmental disorders associated with mutations in the protein-tyrosine phosphatase SHP2. Front Cell Dev Biol 2022; 10:1046415. [PMID: 36407105 PMCID: PMC9672471 DOI: 10.3389/fcell.2022.1046415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Src homology region 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2) is a highly conserved protein tyrosine phosphatase (PTP), which is encoded by PTPN11 and is indispensable during embryonic development. Mutations in PTPN11 in human patients cause aberrant signaling of SHP2, resulting in multiple rare hereditary diseases, including Noonan Syndrome (NS), Noonan Syndrome with Multiple Lentigines (NSML), Juvenile Myelomonocytic Leukemia (JMML) and Metachondromatosis (MC). Somatic mutations in PTPN11 have been found to cause cancer. Here, we focus on the role of SHP2 variants in rare diseases and advances in the understanding of its pathogenesis using model systems.
Collapse
Affiliation(s)
- Maja Solman
- Hubrecht Institute-KNAW, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Jeroen den Hertog
- Hubrecht Institute-KNAW, University Medical Center Utrecht, Utrecht, Netherlands
- Institute Biology Leiden, Leiden University, Leiden, Netherlands
- *Correspondence: Jeroen den Hertog,
| |
Collapse
|
10
|
Tseng CC, Zheng RH, Lin TW, Chou CC, Shih YC, Liang SW, Lee HH. α-Actinin-4 recruits Shp2 into focal adhesions to potentiate ROCK2 activation in podocytes. Life Sci Alliance 2022; 5:5/11/e202201557. [PMID: 36096674 PMCID: PMC9468603 DOI: 10.26508/lsa.202201557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022] Open
Abstract
α-Actinin-4 is crucial in the regulation of Shp2 FA targeting to enhance ROCK2-mediated actomyosin contractility and thereby strengthening cell adhesion and cytoskeletal architecture in podocytes. Cell–matrix adhesions are mainly provided by integrin-mediated focal adhesions (FAs). We previously found that Shp2 is essential for FA maturation by promoting ROCK2 activation at FAs. In this study, we further delineated the role of α-actinin-4 in the FA recruitment and activation of Shp2. We used the conditional immortalized mouse podocytes to examine the role of α-actinin-4 in the regulation of Shp2 and ROCK2 signaling. After the induction of podocyte differentiation, Shp2 and ROCK2 were strongly activated, concomitant with the formation of matured FAs, stress fibers, and interdigitating intracellular junctions in a ROCK-dependent manner. Gene knockout of α-actinin-4 abolished the Shp2 activation and subsequently reduced matured FAs in podocytes. We also demonstrated that gene knockout of ROCK2 impaired the generation of contractility and interdigitating intercellular junctions. Our results reveal the role of α-actinin-4 in the recruitment of Shp2 at FAs to potentiate ROCK2 activation for the maintenance of cellular contractility and cytoskeletal architecture in the cultured podocytes.
Collapse
Affiliation(s)
- Chien-Chun Tseng
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ru-Hsuan Zheng
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Wei Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Chiang Chou
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chia Shih
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shao-Wei Liang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Hui Lee
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan .,Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
11
|
Asmamaw MD, Shi XJ, Zhang LR, Liu HM. A comprehensive review of SHP2 and its role in cancer. Cell Oncol 2022; 45:729-753. [PMID: 36066752 DOI: 10.1007/s13402-022-00698-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 12/26/2022] Open
Abstract
Src homology 2-containing protein tyrosine phosphatase 2 (SHP2) is a non-receptor protein tyrosine phosphatase ubiquitously expressed mainly in the cytoplasm of several tissues. SHP2 modulates diverse cell signaling events that control metabolism, cell growth, differentiation, cell migration, transcription and oncogenic transformation. It interacts with diverse molecules in the cell, and regulates key signaling events including RAS/ERK, PI3K/AKT, JAK/STAT and PD-1 pathways downstream of several receptor tyrosine kinases (RTKs) upon stimulation by growth factors and cytokines. SHP2 acts as both a phosphatase and a scaffold, and plays prominently oncogenic functions but can be tumor suppressor in a context-dependent manner. It typically acts as a positive regulator of RTKs signaling with some inhibitory functions reported as well. SHP2 expression and activity is regulated by such factors as allosteric autoinhibition, microRNAs, ubiquitination and SUMOylation. Dysregulation of SHP2 expression or activity causes many developmental diseases, and hematological and solid tumors. Moreover, upregulated SHP2 expression or activity also decreases sensitivity of cancer cells to anticancer drugs. SHP2 is now considered as a compelling anticancer drug target and several classes of SHP2 inhibitors with different mode of action are developed with some already in clinical trial phases. Moreover, novel SHP2 substrates and functions are rapidly growing both in cell and cancer. In view of this, we comprehensively and thoroughly reviewed literatures about SHP2 regulatory mechanisms, substrates and binding partners, biological functions, roles in human cancers, and different classes of small molecule inhibitors target this oncoprotein in cancer.
Collapse
Affiliation(s)
- Moges Dessale Asmamaw
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory for Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450001, People's Republic of China
| | - Xiao-Jing Shi
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, People's Republic of China
| | - Li-Rong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory for Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450001, People's Republic of China.
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan Province, China. .,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, Henan Province, 450001, People's Republic of China.
| |
Collapse
|
12
|
The Tyrosine Phosphatase SHP2: A New Target for Insulin Resistance? Biomedicines 2022; 10:biomedicines10092139. [PMID: 36140242 PMCID: PMC9495760 DOI: 10.3390/biomedicines10092139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022] Open
Abstract
The SH2 containing protein tyrosine phosphatase 2(SHP2) plays essential roles in fundamental signaling pathways, conferring on it versatile physiological functions during development and in homeostasis maintenance, and leading to major pathological outcomes when dysregulated. Many studies have documented that SHP2 modulation disrupted glucose homeostasis, pointing out a relationship between its dysfunction and insulin resistance, and the therapeutic potential of its targeting. While studies from cellular or tissue-specific models concluded on both pros-and-cons effects of SHP2 on insulin resistance, recent data from integrated systems argued for an insulin resistance promoting role for SHP2, and therefore a therapeutic benefit of its inhibition. In this review, we will summarize the general knowledge of SHP2’s molecular, cellular, and physiological functions, explaining the pathophysiological impact of its dysfunctions, then discuss its protective or promoting roles in insulin resistance as well as the potency and limitations of its pharmacological modulation.
Collapse
|
13
|
Fauser J, Huyot V, Matsche J, Szynal BN, Alexeev Y, Kota P, Karginov AV. Dissecting protein tyrosine phosphatase signaling by engineered chemogenetic control of its activity. J Cell Biol 2022; 221:e202111066. [PMID: 35829702 PMCID: PMC9284425 DOI: 10.1083/jcb.202111066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/06/2022] [Accepted: 06/22/2022] [Indexed: 01/16/2023] Open
Abstract
Protein tyrosine phosphatases (PTPases) are critical mediators of dynamic cell signaling. A tool capable of identifying transient signaling events downstream of PTPases is essential to understand phosphatase function on a physiological time scale. We report a broadly applicable protein engineering method for allosteric regulation of PTPases. This method enables dissection of transient events and reconstruction of individual signaling pathways. Implementation of this approach for Shp2 phosphatase revealed parallel MAPK and ROCK II dependent pathways downstream of Shp2, mediating transient cell spreading and migration. Furthermore, we show that the N-SH2 domain of Shp2 regulates MAPK-independent, ROCK II-dependent cell migration. Engineered targeting of Shp2 activity to different protein complexes revealed that Shp2-FAK signaling induces cell spreading whereas Shp2-Gab1 or Shp2-Gab2 mediates cell migration. We identified specific transient morphodynamic processes induced by Shp2 and determined the role of individual signaling pathways downstream of Shp2 in regulating these events. Broad application of this approach is demonstrated by regulating PTP1B and PTP-PEST phosphatases.
Collapse
Affiliation(s)
- Jordan Fauser
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| | - Vincent Huyot
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| | - Jacob Matsche
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| | - Barbara N. Szynal
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| | | | - Pradeep Kota
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Andrei V. Karginov
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
14
|
Marasco M, Kirkpatrick J, Nanna V, Sikorska J, Carlomagno T. Phosphotyrosine couples peptide binding and SHP2 activation via a dynamic allosteric network. Comput Struct Biotechnol J 2021; 19:2398-2415. [PMID: 34025932 PMCID: PMC8113834 DOI: 10.1016/j.csbj.2021.04.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 11/18/2022] Open
Abstract
SHP2 is a ubiquitous protein tyrosine phosphatase, whose activity is regulated by phosphotyrosine (pY)-containing peptides generated in response to extracellular stimuli. Its crystal structure reveals a closed, auto-inhibited conformation in which the N-terminal Src homology 2 (N-SH2) domain occludes the catalytic site of the phosphatase (PTP) domain. High-affinity mono-phosphorylated peptides promote catalytic activity by binding to N-SH2 and disrupting the interaction with the PTP. The mechanism behind this process is not entirely clear, especially because N-SH2 is incapable of accommodating complete peptide binding when SHP2 is in the auto-inhibited state. Here, we show that pY performs an essential role in this process; in addition to its contribution to overall peptide-binding energy, pY-recognition leads to enhanced dynamics of the N-SH2 EF and BG loops via an allosteric communication network, which destabilizes the N-SH2-PTP interaction surface and simultaneously generates a fully accessible binding pocket for the C-terminal half of the phosphopeptide. Subsequently, full binding of the phosphopeptide is associated with the stabilization of activated SHP2. We demonstrate that this allosteric network exists only in N-SH2, which is directly involved in the regulation of SHP2 activity, while the C-terminal SH2 domain (C-SH2) functions primarily to recruit high-affinity bidentate phosphopeptides.
Collapse
Affiliation(s)
- Michelangelo Marasco
- Leibniz University Hannover, Center of Biomolecular Drug Research and Institute of Organic Chemistry, Schneiderberg 38, 30167 Hannover, Germany
| | - John Kirkpatrick
- Leibniz University Hannover, Center of Biomolecular Drug Research and Institute of Organic Chemistry, Schneiderberg 38, 30167 Hannover, Germany
- Helmholtz Center for Infection Research, Group of NMR-based Structural Chemistry, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Vittoria Nanna
- Leibniz University Hannover, Center of Biomolecular Drug Research and Institute of Organic Chemistry, Schneiderberg 38, 30167 Hannover, Germany
| | - Justyna Sikorska
- Helmholtz Center for Infection Research, Group of NMR-based Structural Chemistry, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Teresa Carlomagno
- Leibniz University Hannover, Center of Biomolecular Drug Research and Institute of Organic Chemistry, Schneiderberg 38, 30167 Hannover, Germany
- Helmholtz Center for Infection Research, Group of NMR-based Structural Chemistry, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
15
|
Lin CC, Wieteska L, Suen KM, Kalverda AP, Ahmed Z, Ladbury JE. Grb2 binding induces phosphorylation-independent activation of Shp2. Commun Biol 2021; 4:437. [PMID: 33795832 PMCID: PMC8016844 DOI: 10.1038/s42003-021-01969-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/25/2021] [Indexed: 11/12/2022] Open
Abstract
The regulation of phosphatase activity is fundamental to the control of intracellular signalling and in particular the tyrosine kinase-mediated mitogen-activated protein kinase (MAPK) pathway. Shp2 is a ubiquitously expressed protein tyrosine phosphatase and its kinase-induced hyperactivity is associated with many cancer types. In non-stimulated cells we find that binding of the adaptor protein Grb2, in its monomeric state, initiates Shp2 activity independent of phosphatase phosphorylation. Grb2 forms a bidentate interaction with both the N-terminal SH2 and the catalytic domains of Shp2, releasing the phosphatase from its auto-inhibited conformation. Grb2 typically exists as a dimer in the cytoplasm. However, its monomeric state prevails under basal conditions when it is expressed at low concentration, or when it is constitutively phosphorylated on a specific tyrosine residue (Y160). Thus, Grb2 can activate Shp2 and downstream signal transduction, in the absence of extracellular growth factor stimulation or kinase-activating mutations, in response to defined cellular conditions. Therefore, direct binding of Grb2 activates Shp2 phosphatase in the absence of receptor tyrosine kinase up-regulation.
Collapse
Affiliation(s)
- Chi-Chuan Lin
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| | - Lukasz Wieteska
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Kin Man Suen
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Arnout P Kalverda
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Zamal Ahmed
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John E Ladbury
- School of Molecular and Cellular Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India.
| |
Collapse
|
16
|
Kerr DL, Haderk F, Bivona TG. Allosteric SHP2 inhibitors in cancer: Targeting the intersection of RAS, resistance, and the immune microenvironment. Curr Opin Chem Biol 2021; 62:1-12. [PMID: 33418513 DOI: 10.1016/j.cbpa.2020.11.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/09/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023]
Abstract
The nonreceptor protein tyrosine phosphatase SHP2 (encoded by PTPN11) integrates growth and differentiation signals from receptor tyrosine kinases (RTKs) into the RAS/mitogen-activated protein kinase (MAPK) cascade. Considered 'undruggable' over three decades, SHP2 is now a potentially druggable target with the advent of allosteric SHP2 inhibitors. These agents hold promise for improving patient outcomes, showing efficacy in preclinical cancer models, where SHP2 is critical for either oncogenic signaling or resistance to current targeted agents. SHP2 inhibition may also produce immunomodulatory effects in certain tumor microenvironment cells to help cultivate antitumor immune responses. The first generation of allosteric SHP2 inhibitors is under clinical evaluation to determine safety, appropriate tolerability management, and antitumor efficacy, investigations that will dictate future clinical applications.
Collapse
Affiliation(s)
- D Lucas Kerr
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Franziska Haderk
- Department of Medicine, University of California, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| |
Collapse
|
17
|
Chen H, Libring S, Ruddraraju KV, Miao J, Solorio L, Zhang ZY, Wendt MK. SHP2 is a multifunctional therapeutic target in drug resistant metastatic breast cancer. Oncogene 2020; 39:7166-7180. [PMID: 33033382 PMCID: PMC7714690 DOI: 10.1038/s41388-020-01488-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 01/08/2023]
Abstract
Metastatic breast cancer (MBC) is an extremely recalcitrant disease capable of bypassing current targeted therapies via engagement of several growth promoting pathways. SH2 containing protein tyrosine phosphatase-2 (SHP2) is an oncogenic phosphatase known to facilitate growth and survival signaling downstream of numerous receptor inputs. Herein, we used inducible genetic depletion and two distinct pharmacological inhibitors to investigate the therapeutic potential of targeting SHP2 in MBC. Cells that acquired resistance to the ErbB kinase inhibitor, neratinib, displayed increased phosphorylation of SHP2 at the Y542 activation site. In addition, higher levels of SHP2 phosphorylation, but not expression, were associated with decreased survival of breast cancer patients. Pharmacological inhibition of SHP2 activity blocked ERK1/2 and AKT signaling generated from exogenous stimulation with FGF2, PDGF, and hGF and readily prevented MBC cell growth induced by these factors. SHP2 was also phosphorylated upon engagement of the extracellular matrix (ECM) via focal adhesion kinase. Consistent with the potential of SHP2-targeted compounds as therapeutic agents, the growth inhibitory property of SHP2 blockade was enhanced in ECM-rich 3D culture environments. In vivo blockade of SHP2 in the adjuvant setting decreased pulmonary metastasis and extended the survival of systemic tumor-bearing mice. Finally, inhibition of SHP2 in combination with FGFR-targeted kinase inhibitors synergistically blocked the growth of MBC cells. Overall, our findings support the conclusion that SHP2 constitutes a shared signaling node allowing MBC cells to simultaneously engage a diversity of growth and survival pathways, including those derived from the ECM.
Collapse
Affiliation(s)
- Hao Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Sarah Libring
- Department of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | | | - Jinmin Miao
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Luis Solorio
- Department of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Michael K Wendt
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
18
|
Zhu D, Wu P, Xiao C, Hu W, Zhang T, Hu X, Chen W, Wang J. Inflammatory Cytokines Alter Mesenchymal Stem Cell Mechanosensing and Adhesion on Stiffened Infarct Heart Tissue After Myocardial Infarction. Front Cell Dev Biol 2020; 8:583700. [PMID: 33195229 PMCID: PMC7645114 DOI: 10.3389/fcell.2020.583700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has demonstrated its potential in repairing infarct heart tissue and recovering heart function after myocardial infarction (MI). However, its therapeutic effect is still limited due to poor MSC engraftment at the injury site whose tissue stiffness and local inflammation both dynamically and rapidly change after MI. Whether and how inflammatory cytokines could couple with stiffness change to affect MSC engraftment in the infarct zone still remain unclear. In this study, we characterized dynamic stiffness changes of and inflammatory cytokine expression in the infarct region of rat heart within a month after MI. We found that the tissue stiffness of the heart tissue gradually increased and peaked 21 days after MI along with the rapid upregulation of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) in the first 3 days, followed by a sharp decline. We further demonstrated in vitro that immobilized inflammatory cytokine IL-6 performed better than the soluble form in enhancing MSC adhesion to stiffened substrate through IL-6/src homology 2 (SH2) domain-containing tyrosine phosphatase-2 (SHP2)/integrin signaling axis. We also confirmed such mechano-immune coupling of tissue stiffness and inflammatory cytokines in modulating MSC engraftment in the rat heart after MI in vivo. Our study provides new mechanistic insights of mechanical–inflammation coupling to improve MSC mechanosensing and adhesion, potentially benefiting MSC engraftment and its clinical therapy for MI.
Collapse
Affiliation(s)
- Dan Zhu
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Wu
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Changchen Xiao
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Hu
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Tongtong Zhang
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Xinyang Hu
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Chen
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Jian'an Wang
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
19
|
Patsoukis N, Duke-Cohan JS, Chaudhri A, Aksoylar HI, Wang Q, Council A, Berg A, Freeman GJ, Boussiotis VA. Interaction of SHP-2 SH2 domains with PD-1 ITSM induces PD-1 dimerization and SHP-2 activation. Commun Biol 2020; 3:128. [PMID: 32184441 PMCID: PMC7078208 DOI: 10.1038/s42003-020-0845-0] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
Programmed cell death-1 (PD-1) inhibits T cell responses. This function relies on interaction with SHP-2. PD-1 has one immunoreceptor tyrosine-based inhibitory motif (ITIM) at Y223 and one immunoreceptor tyrosine-based switch motif (ITSM) at Y248. Only ITSM-Y248 is indispensable for PD-1-mediated inhibitory function but how SHP-2 enzymatic activation is mechanistically regulated by one PD-1 phosphotyrosine remains a puzzle. We found that after PD-1 phosphorylation, SHP-2 can bridge phosphorylated ITSM-Y248 residues on two PD-1 molecules via its amino terminal (N)-SH2 and carboxyterminal (C)-SH2 domains forming a PD-1: PD-1 dimer in live cells. The biophysical ability of SHP-2 to interact with two ITSM-pY248 residues was documented by isothermal titration calorimetry. SHP-2 interaction with two ITSM-pY248 phosphopeptides induced robust enzymatic activation. Our results unravel a mechanism of PD-1: SHP-2 interaction that depends only on ITSM-Y248 and explain how a single docking site within the PD-1 cytoplasmic tail can activate SHP-2 and PD-1-mediated inhibitory function.
Collapse
Affiliation(s)
- Nikolaos Patsoukis
- Division of Hematology-Oncology Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Jonathan S Duke-Cohan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Apoorvi Chaudhri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Halil-Ibrahim Aksoylar
- Division of Hematology-Oncology Beth Israel Deaconess Medical Center, 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, Boston, MA, 02215, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Asia Council
- Division of Hematology-Oncology Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Anders Berg
- Department of Pathology Beth Israel Deaconess Cancer Center, Harvard Medical School, Boston, MA, 02215, USA
- Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
20
|
Marasco M, Berteotti A, Weyershaeuser J, Thorausch N, Sikorska J, Krausze J, Brandt HJ, Kirkpatrick J, Rios P, Schamel WW, Köhn M, Carlomagno T. Molecular mechanism of SHP2 activation by PD-1 stimulation. SCIENCE ADVANCES 2020; 6:eaay4458. [PMID: 32064351 PMCID: PMC6994217 DOI: 10.1126/sciadv.aay4458] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/22/2019] [Indexed: 05/02/2023]
Abstract
In cancer, the programmed death-1 (PD-1) pathway suppresses T cell stimulation and mediates immune escape. Upon stimulation, PD-1 becomes phosphorylated at its immune receptor tyrosine-based inhibitory motif (ITIM) and immune receptor tyrosine-based switch motif (ITSM), which then bind the Src homology 2 (SH2) domains of SH2-containing phosphatase 2 (SHP2), initiating T cell inactivation. The SHP2-PD-1 complex structure and the exact functions of the two SH2 domains and phosphorylated motifs remain unknown. Here, we explain the structural basis and provide functional evidence for the mechanism of PD-1-mediated SHP2 activation. We demonstrate that full activation is obtained only upon phosphorylation of both ITIM and ITSM: ITSM binds C-SH2 with strong affinity, recruiting SHP2 to PD-1, while ITIM binds N-SH2, displacing it from the catalytic pocket and activating SHP2. This binding event requires the formation of a new inter-domain interface, offering opportunities for the development of novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- M. Marasco
- Leibniz University Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research, Schneiderberg 38, 30167 Hannover, Germany
| | - A. Berteotti
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - J. Weyershaeuser
- Faculty of Biology, Institute of Biology III, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - N. Thorausch
- Faculty of Biology, Institute of Biology III, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - J. Sikorska
- Helmholtz Centre for Infection Research, Group of Structural Chemistry, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - J. Krausze
- Leibniz University Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research, Schneiderberg 38, 30167 Hannover, Germany
| | - H. J. Brandt
- Faculty of Biology, Institute of Biology III, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - J. Kirkpatrick
- Leibniz University Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research, Schneiderberg 38, 30167 Hannover, Germany
- Helmholtz Centre for Infection Research, Group of Structural Chemistry, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - P. Rios
- European Molecular Biology Laboratory, Genome Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
- Faculty of Biology, Institute of Biology III, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - W. W. Schamel
- Faculty of Biology, Institute of Biology III, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - M. Köhn
- European Molecular Biology Laboratory, Genome Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
- Faculty of Biology, Institute of Biology III, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Corresponding author. (T.C.); (M.K.)
| | - T. Carlomagno
- Leibniz University Hannover, Institute of Organic Chemistry and Center for Biomolecular Drug Research, Schneiderberg 38, 30167 Hannover, Germany
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
- Helmholtz Centre for Infection Research, Group of Structural Chemistry, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- Corresponding author. (T.C.); (M.K.)
| |
Collapse
|
21
|
Deubiquitinase PSMD14 enhances hepatocellular carcinoma growth and metastasis by stabilizing GRB2. Cancer Lett 2020; 469:22-34. [DOI: 10.1016/j.canlet.2019.10.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/23/2019] [Accepted: 10/15/2019] [Indexed: 01/17/2023]
|
22
|
Qin Q, Laub S, Shi Y, Ouyang M, Peng Q, Zhang J, Wang Y, Lu S. Fluocell for Ratiometric and High-Throughput Live-Cell Image Visualization and Quantitation. FRONTIERS IN PHYSICS 2019; 7:154. [PMID: 33163483 PMCID: PMC7646842 DOI: 10.3389/fphy.2019.00154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Spatiotemporal regulation of molecular activities dictates cellular function and fate. Investigation of dynamic molecular activities in live cells often requires the visualization and quantitation of fluorescent ratio image sequences with subcellular resolution and in high throughput. Hence, there is a great need for convenient software tools specifically designed with these capabilities. Here we describe a well-characterized open-source software package, Fluocell, customized to visualize pixelwise ratiometric images and calculate ratio time courses with subcellular resolution and in high throughput. Fluocell also provides group statistics and kinetic analysis functions for the quantified time courses, as well as 3D structure and function visualization for ratio images. The application of Fluocell is demonstrated by the ratiometric analysis of intensity images for several single-chain Förster (or fluorescence) resonance energy transfer (FRET)-based biosensors, allowing efficient quantification of dynamic molecular activities in a heterogeneous population of single live cells. Our analysis revealed distinct activation kinetics of Fyn kinase in the cytosolic and membrane compartments, and visualized a 4D spatiotemporal distribution of epigenetic signals in mitotic cells. Therefore, Fluocell provides an integrated environment for ratiometric live-cell image visualization and analysis, which generates high-quality single-cell dynamic data and allows the quantitative machine-learning of biophysical and biochemical computational models for molecular regulations in cells and tissues.
Collapse
Affiliation(s)
- Qin Qin
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Shannon Laub
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Yiwen Shi
- Department of Mathematics, Center of Computational Mathematics, University of California, San Diego, San Diego, CA, United State
| | - Mingxing Ouyang
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, China
| | - Qin Peng
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Shaoying Lu
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
- Department of Mathematics, Center of Computational Mathematics, University of California, San Diego, San Diego, CA, United State
| |
Collapse
|
23
|
Ryu HH, Kim T, Kim JW, Kang M, Park P, Kim YG, Kim H, Ha J, Choi JE, Lee J, Lim CS, Kim CH, Kim SJ, Silva AJ, Kaang BK, Lee YS. Excitatory neuron-specific SHP2-ERK signaling network regulates synaptic plasticity and memory. Sci Signal 2019; 12:12/571/eaau5755. [PMID: 30837304 DOI: 10.1126/scisignal.aau5755] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mutations in RAS signaling pathway components cause diverse neurodevelopmental disorders, collectively called RASopathies. Previous studies have suggested that dysregulation in RAS-extracellular signal-regulated kinase (ERK) activation is restricted to distinct cell types in different RASopathies. Some cases of Noonan syndrome (NS) are associated with gain-of-function mutations in the phosphatase SHP2 (encoded by PTPN11); however, SHP2 is abundant in multiple cell types, so it is unclear which cell type(s) contribute to NS phenotypes. Here, we found that expressing the NS-associated mutant SHP2D61G in excitatory, but not inhibitory, hippocampal neurons increased ERK signaling and impaired both long-term potentiation (LTP) and spatial memory in mice, although endogenous SHP2 was expressed in both neuronal types. Transcriptomic analyses revealed that the genes encoding SHP2-interacting proteins that are critical for ERK activation, such as GAB1 and GRB2, were enriched in excitatory neurons. Accordingly, expressing a dominant-negative mutant of GAB1, which reduced its interaction with SHP2D61G, selectively in excitatory neurons, reversed SHP2D61G-mediated deficits. Moreover, ectopic expression of GAB1 and GRB2 together with SHP2D61G in inhibitory neurons resulted in ERK activation. These results demonstrate that RAS-ERK signaling networks are notably different between excitatory and inhibitory neurons, accounting for the cell type-specific pathophysiology of NS and perhaps other RASopathies.
Collapse
Affiliation(s)
- Hyun-Hee Ryu
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - TaeHyun Kim
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Minkyung Kang
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Pojeong Park
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Yong Gyu Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyopil Kim
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Jiyeon Ha
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ja Eun Choi
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Jisu Lee
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Chae-Seok Lim
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan 54538, Korea
| | - Chul-Hong Kim
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Alcino J Silva
- Department of Neurobiology, Integrative Center for Learning and Memory, Brain Research Institute, University of California Los Angeles, California, CA 90095, USA
| | - Bong-Kiun Kaang
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea. .,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
24
|
Bredrup C, Stokowy T, McGaughran J, Lee S, Sapkota D, Cristea I, Xu L, Tveit KS, Høvding G, Steen VM, Rødahl E, Bruland O, Houge G. A tyrosine kinase-activating variant Asn666Ser in PDGFRB causes a progeria-like condition in the severe end of Penttinen syndrome. Eur J Hum Genet 2018; 27:574-581. [PMID: 30573803 DOI: 10.1038/s41431-018-0323-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/02/2018] [Accepted: 11/07/2018] [Indexed: 01/08/2023] Open
Abstract
Missense variants located to the "molecular brake" in the tyrosine kinase hinge region of platelet-derived growth factor receptor-β, encoded by PFGFRB, can cause Penttinen-type (Val665Ala) and Penttinen-like (Asn666His) premature ageing syndromes, as well as infantile myofibromatosis (Asn666Lys and Pro660Thr). We have found the same de novo PDGFRB c.1997A>G p.(Asn666Ser) variants in two patients with lipodystrophy, acro-osteolysis and severely reduced vision due to corneal neovascularisation, reminiscent of a severe form of Penttinen syndrome with more pronounced connective tissue destruction. In line with this phenotype, patient skin fibroblasts were prone to apoptosis. Both in patient fibroblasts and stably transduced HeLa and HEK293 cells, autophosphorylation of PDGFRβ was observed, as well as increased phosphorylation of downstream signalling proteins such as STAT1, PLCγ1, PTPN11/SHP2-Tyr580 and AKT. Phosphorylation of MAPK3 (ERK1) and PTPN11/SHP2-Tyr542 appeared unaffected. This suggests that this missense change not only weakens tyrosine kinase autoinhibition, but also influences substrate binding, as both PTPN11 tyrosines (Tyr542 and Tyr580) usually are phosphorylated upon PDGFR activation. Imatinib was a strong inhibitor of phosphorylation of all these targets, suggesting an option for precision medicine based treatment.
Collapse
Affiliation(s)
- Cecilie Bredrup
- Department of Medical Genetics, Haukeland University Hospital, 5021, Bergen, Norway.,Department of Ophthalmology, Haukeland University Hospital, 5021, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, 5020, Bergen, Norway
| | - Tomasz Stokowy
- Department of Medical Genetics, Haukeland University Hospital, 5021, Bergen, Norway.,Department of Clinical Science, University of Bergen, 5020, Bergen, Norway
| | - Julie McGaughran
- Genetic Health QLD, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Samuel Lee
- Genetic Health QLD, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Dipak Sapkota
- Department of Oncology and Medical Physics, Haukeland University Hospital, 5021, Bergen, Norway.,Institute of Oral Biology, University of Oslo, 0315, Oslo, Norway
| | - Ileana Cristea
- Department of Clinical Medicine, University of Bergen, 5020, Bergen, Norway
| | - Linda Xu
- Department of Clinical Medicine, University of Bergen, 5020, Bergen, Norway
| | - Kåre Steinar Tveit
- Department of Dermatology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Gunnar Høvding
- Department of Ophthalmology, Haukeland University Hospital, 5021, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, 5020, Bergen, Norway
| | - Vidar Martin Steen
- Department of Medical Genetics, Haukeland University Hospital, 5021, Bergen, Norway.,Department of Clinical Science, University of Bergen, 5020, Bergen, Norway
| | - Eyvind Rødahl
- Department of Ophthalmology, Haukeland University Hospital, 5021, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, 5020, Bergen, Norway
| | - Ove Bruland
- Department of Medical Genetics, Haukeland University Hospital, 5021, Bergen, Norway
| | - Gunnar Houge
- Department of Medical Genetics, Haukeland University Hospital, 5021, Bergen, Norway.
| |
Collapse
|
25
|
Peled M, Strazza M, Mor A. Co-immunoprecipitation Assay for Studying Functional Interactions Between Receptors and Enzymes. J Vis Exp 2018. [PMID: 30320758 DOI: 10.3791/58433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Receptor-associated enzymes are the major mediators of cellular activation. These enzymes are regulated, at least in part, by physical interactions with cytoplasmic tails of the receptors. The interactions often occur through specific protein domains and result in activation of the enzymes. There are several methods to study interactions between proteins. While co-immunoprecipitation is commonly used to study domains that are required for protein-protein interactions, there are no assays that document the contribution of specific domains to activity of the recruited enzymes at the same time. Accordingly, the method described here combines co-immunoprecipitation and an on-bead enzymatic activity assay for simultaneous evaluation of interactions between proteins and associated enzymatic activation. The goal of this protocol is to identify the domains that are critical for physical interactions between a protein and enzyme and the domains that are obligatory for complete activation of the enzyme. The importance of this assay is demonstrated, as certain receptor protein domains contribute to the binding of the enzyme to the cytoplasmic tail of the receptor, while other domains are necessary to regulate the function of the same enzyme.
Collapse
Affiliation(s)
- Michael Peled
- Pulmonary Department, The Chaim Sheba Medical Center
| | - Marianne Strazza
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center
| | - Adam Mor
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center;
| |
Collapse
|
26
|
Hale AJ, den Hertog J. Shp2-Mitogen-Activated Protein Kinase Signaling Drives Proliferation during Zebrafish Embryo Caudal Fin Fold Regeneration. Mol Cell Biol 2018; 38:e00515-17. [PMID: 29203641 PMCID: PMC5789028 DOI: 10.1128/mcb.00515-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 10/18/2017] [Accepted: 11/23/2017] [Indexed: 11/25/2022] Open
Abstract
Regeneration of the zebrafish caudal fin following amputation occurs through wound healing, followed by formation of a blastema, which produces cells to replace the lost tissue in the final phase of regenerative outgrowth. We show that ptpn11a-/- ptpn11b-/- zebrafish embryos, lacking functional Shp2, fail to regenerate their caudal fin folds. Rescue experiments indicated that Shp2a has a functional signaling role, requiring its catalytic activity and SH2 domains but not the two C-terminal tyrosine phosphorylation sites. Surprisingly, expression of Shp2a variants with increased and reduced catalytic activity, respectively, rescued caudal fin fold regeneration to similar extents. Expression of mmp9 and junbb, indicative of formation of the wound epidermis and distal blastema, respectively, suggested that these processes occurred in ptpn11a-/- ptpn11b-/- zebrafish embryos. However, cell proliferation and MAPK phosphorylation were reduced. Pharmacological inhibition of MEK1 in wild-type zebrafish embryos phenocopied loss of Shp2. Our results suggest an essential role for Shp2a-mitogen-activated protein kinase (MAPK) signaling in promoting cell proliferation during zebrafish embryo caudal fin fold regeneration.
Collapse
Affiliation(s)
- Alexander James Hale
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
- Institute Biology Leiden, Leiden University, Leiden, the Netherlands
| | - Jeroen den Hertog
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
- Institute Biology Leiden, Leiden University, Leiden, the Netherlands
| |
Collapse
|
27
|
Affinity purification mass spectrometry analysis of PD-1 uncovers SAP as a new checkpoint inhibitor. Proc Natl Acad Sci U S A 2017; 115:E468-E477. [PMID: 29282323 DOI: 10.1073/pnas.1710437115] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Programmed cell death-1 (PD-1) is an essential inhibitory receptor in T cells. Antibodies targeting PD-1 elicit durable clinical responses in patients with multiple tumor indications. Nevertheless, a significant proportion of patients do not respond to anti-PD-1 treatment, and a better understanding of the signaling pathways downstream of PD-1 could provide biomarkers for those whose tumors respond and new therapeutic approaches for those whose tumors do not. We used affinity purification mass spectrometry to uncover multiple proteins associated with PD-1. Among these proteins, signaling lymphocytic activation molecule-associated protein (SAP) was functionally and mechanistically analyzed for its contribution to PD-1 inhibitory responses. Silencing of SAP augmented and overexpression blocked PD-1 function. T cells from patients with X-linked lymphoproliferative disease (XLP), who lack functional SAP, were hyperresponsive to PD-1 signaling, confirming its inhibitory role downstream of PD-1. Strikingly, signaling downstream of PD-1 in purified T cell subsets did not correlate with PD-1 surface expression but was inversely correlated with intracellular SAP levels. Mechanistically, SAP opposed PD-1 function by acting as a molecular shield of key tyrosine residues that are targets for the tyrosine phosphatase SHP2, which mediates PD-1 inhibitory properties. Our results identify SAP as an inhibitor of PD-1 function and SHP2 as a potential therapeutic target in patients with XLP.
Collapse
|
28
|
Transancestral mapping and genetic load in systemic lupus erythematosus. Nat Commun 2017; 8:16021. [PMID: 28714469 PMCID: PMC5520018 DOI: 10.1038/ncomms16021] [Citation(s) in RCA: 283] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 05/23/2017] [Indexed: 12/27/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with marked gender and ethnic disparities. We report a large transancestral association study of SLE using Immunochip genotype data from 27,574 individuals of European (EA), African (AA) and Hispanic Amerindian (HA) ancestry. We identify 58 distinct non-HLA regions in EA, 9 in AA and 16 in HA (∼50% of these regions have multiple independent associations); these include 24 novel SLE regions (P<5 × 10−8), refined association signals in established regions, extended associations to additional ancestries, and a disentangled complex HLA multigenic effect. The risk allele count (genetic load) exhibits an accelerating pattern of SLE risk, leading us to posit a cumulative hit hypothesis for autoimmune disease. Comparing results across the three ancestries identifies both ancestry-dependent and ancestry-independent contributions to SLE risk. Our results are consistent with the unique and complex histories of the populations sampled, and collectively help clarify the genetic architecture and ethnic disparities in SLE. Systemic lupus erythematosus (SLE) is an autoimmune disease with a strong ethnic and gender bias. In a transancestral genetic association study, Langefeld et al. identify 24 novel regions associated with risk to lupus and propose a cumulative hits hypothesis for loci conferring risk to SLE.
Collapse
|
29
|
SHP2 sails from physiology to pathology. Eur J Med Genet 2015; 58:509-25. [PMID: 26341048 DOI: 10.1016/j.ejmg.2015.08.005] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/24/2015] [Accepted: 08/30/2015] [Indexed: 02/08/2023]
Abstract
Over the two past decades, mutations of the PTPN11 gene, encoding the ubiquitous protein tyrosine phosphatase SHP2 (SH2 domain-containing tyrosine phosphatase 2), have been identified as the causal factor of several developmental diseases (Noonan syndrome (NS), Noonan syndrome with multiple lentigines (NS-ML), and metachondromatosis), and malignancies (juvenile myelomonocytic leukemia). SHP2 plays essential physiological functions in organism development and homeostasis maintenance by regulating fundamental intracellular signaling pathways in response to a wide range of growth factors and hormones, notably the pleiotropic Ras/Mitogen-Activated Protein Kinase (MAPK) and the Phosphoinositide-3 Kinase (PI3K)/AKT cascades. Analysis of the biochemical impacts of PTPN11 mutations first identified both loss-of-function and gain-of-function mutations, as well as more subtle defects, highlighting the major pathophysiological consequences of SHP2 dysregulation. Then, functional genetic studies provided insights into the molecular dysregulations that link SHP2 mutants to the development of specific traits of the diseases, paving the way for the design of specific therapies for affected patients. In this review, we first provide an overview of SHP2's structure and regulation, then describe its molecular roles, notably its functions in modulating the Ras/MAPK and PI3K/AKT signaling pathways, and its physiological roles in organism development and homeostasis. In the second part, we describe the different PTPN11 mutation-associated pathologies and their clinical manifestations, with particular focus on the biochemical and signaling outcomes of NS and NS-ML-associated mutations, and on the recent advances regarding the pathophysiology of these diseases.
Collapse
|
30
|
Sun J, Jakobsson E, Wang Y, Brinker CJ. Nanoporous silica-based protocells at multiple scales for designs of life and nanomedicine. Life (Basel) 2015; 5:214-29. [PMID: 25607812 PMCID: PMC4390849 DOI: 10.3390/life5010214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/29/2014] [Accepted: 01/06/2015] [Indexed: 11/16/2022] Open
Abstract
Various protocell models have been constructed de novo with the bottom-up approach. Here we describe a silica-based protocell composed of a nanoporous amorphous silica core encapsulated within a lipid bilayer built by self-assembly that provides for independent definition of cell interior and the surface membrane. In this review, we will first describe the essential features of this architecture and then summarize the current development of silica-based protocells at both micro- and nanoscale with diverse functionalities. As the structure of the silica is relatively static, silica-core protocells do not have the ability to change shape, but their interior structure provides a highly crowded and, in some cases, authentic scaffold upon which biomolecular components and systems could be reconstituted. In basic research, the larger protocells based on precise silica replicas of cells could be developed into geometrically realistic bioreactor platforms to enable cellular functions like coupled biochemical reactions, while in translational research smaller protocells based on mesoporous silica nanoparticles are being developed for targeted nanomedicine. Ultimately we see two different motivations for protocell research and development: (1) to emulate life in order to understand it; and (2) to use biomimicry to engineer desired cellular interactions.
Collapse
Affiliation(s)
- Jie Sun
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Eric Jakobsson
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yingxiao Wang
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - C Jeffrey Brinker
- Department of Chemical and Nuclear Engineering, Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NW 87106, USA.
| |
Collapse
|
31
|
Lu S, Seong J, Wang Y, Chang SC, Eichorst JP, Ouyang M, Li JYS, Chien S, Wang Y. Decipher the dynamic coordination between enzymatic activity and structural modulation at focal adhesions in living cells. Sci Rep 2014; 4:5756. [PMID: 25056908 PMCID: PMC4108961 DOI: 10.1038/srep05756] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 06/23/2014] [Indexed: 02/06/2023] Open
Abstract
Focal adhesions (FAs) are dynamic subcellular structures crucial for cell adhesion, migration and differentiation. It remains an enigma how enzymatic activities in these local complexes regulate their structural remodeling in live cells. Utilizing biosensors based on fluorescence resonance energy transfer (FRET), we developed a correlative FRET imaging microscopy (CFIM) approach to quantitatively analyze the subcellular coordination between the enzymatic Src activation and the structural FA disassembly. CFIM reveals that the Src kinase activity only within the microdomain of lipid rafts at the plasma membrane is coupled with FA dynamics. FA disassembly at cell periphery was linearly dependent on this raft-localized Src activity, although cells displayed heterogeneous levels of response to stimulation. Within lipid rafts, the time delay between Src activation and FA disassembly was 1.2 min in cells seeded on low fibronectin concentration ([FN]) and 4.3 min in cells on high [FN]. CFIM further showed that the level of Src-FA coupling, as well as the time delay, was regulated by cell-matrix interactions, as a tight enzyme-structure coupling occurred in FA populations mediated by integrin αvβ₃, but not in those by integrin α₅β₁. Therefore, different FA subpopulations have distinctive regulation mechanisms between their local kinase activity and structural FA dynamics.
Collapse
Affiliation(s)
- Shaoying Lu
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093-0435
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Jihye Seong
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Current address: Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Yi Wang
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Shiou-chi Chang
- Department of Chemical Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - John Paul Eichorst
- Center of Biophysics and Computational Biology, Beckman Institute for Advanced Science and Technology, Department of Molecular and Integrative Physiology and, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Mingxing Ouyang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093-0435
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Julie Y.-S. Li
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093-0435
| | - Shu Chien
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093-0435
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093-0435
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Center of Biophysics and Computational Biology, Beckman Institute for Advanced Science and Technology, Department of Molecular and Integrative Physiology and, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
32
|
Qu Y, Chen Q, Lai X, Zhu C, Chen C, Zhao X, Deng R, Xu M, Yuan H, Wang Y, Yu J, Huang J. SUMOylation of Grb2 enhances the ERK activity by increasing its binding with Sos1. Mol Cancer 2014; 13:95. [PMID: 24775912 PMCID: PMC4021559 DOI: 10.1186/1476-4598-13-95] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 04/21/2014] [Indexed: 02/02/2023] Open
Abstract
Background Grb2 (Growth factor receptor-bound protein 2) is a key adaptor protein in maintaining the ERK activity via linking Sos1 (Son of sevenless homolog 1) or other proteins to activated RTKs, such as EGFR. Currently, little knowledge is available concerning the post-translational modification (PTM) of Grb2 except for its phosphorylation. Since emerging evidences have highlighted the importance of SUMOylation (Small ubiquitin-related modifier), a reversible PTM, in modulating protein functions, we wondered if Grb2 could be SUMOylated and thereby influences its functions especially involved in the Ras/MEK/ERK pathway. Methods SUMOylation of Grb2 was analyzed with the in vivo SUMOylation assay using the Ni2+-NTA affinity pulldown and the in vitro E.coli-based SUMOylation assay. To test the ERK activity and cell transformation, the murine fibroblast cell line NIH/3T3 and the murine colon cancer cell line CMT-93 were used for the experiments including Grb2 knockdown, ectopic re-expression, cell transformation and migration. Immunoprecipitation (IP) was employed for seeking proteins that interact with SUMO modified Grb2. Xenograft tumor model in mice was conducted to verify that Grb2 SUMOylation regulated tumorigenesis in vivo. Results Grb2 can be SUMOylated by SUMO1 at lysine 56 (K56), which is located in the linker region between the N-terminal SH3 domain and the SH2 domain. Knockdown of Grb2 reduced the ERK activity and suppressed cell motility and tumorigenesis in vitro and in vivo, which were all rescued by stable ectopic re-expression of wild-type Grb2 but not the mutant Grb2K56R. Furthermore, Grb2 SUMOylation at K56 increased the formation of Grb2-Sos1 complex, which sequentially leads to the activation of Ras/MEK/MAPK pathway. Conclusions Our results provide evidences that Grb2 is SUMOylated in vivo and this modification enhances ERK activities via increasing the formation of Grb2-Sos1 complex, and may consequently promote cell motility, transformation and tumorigenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China.
| | | |
Collapse
|