1
|
Xue N, Zhao J, Yin J, Liu L, Yang Z, Zhai S, Bian X, Gao X. The Role of SUMO1 Modification of SOX9 in Cartilage Development Stimulated by Zinc Ions in Mice. Organogenesis 2025; 21:2460269. [PMID: 39905673 PMCID: PMC11801356 DOI: 10.1080/15476278.2025.2460269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/09/2024] [Accepted: 01/25/2025] [Indexed: 02/06/2025] Open
Abstract
Zinc ions play a pivotal role in facilitating the development of cartilage in mice. Nevertheless, the precise underlying mechanism remains elusive. Our investigation was centered on elucidating the impact of zinc deficiency on cartilage maturation by modulating SUMO1 and UBC9 at both the protein and mRNA levels. We administered a regimen inducing zinc deficiency to gravid mice from E0.5 until euthanasia. Subsequently, we subjected the embryos to scrutiny employing HE, Safranin O staining and IHC. Primary chondrocytes were isolated from fetal mouse femoral condyles and utilized for Western blot analysis to discern the expression profiles of SUMO1, SUMO2/3, UBC9, SOX9, MMP13, Collagen II, RUNX2, and aggrecan. Furthermore, ATDC5 murine chondrocytes were subjected to treatment with ZnCl2, followed by RT-PCR assessment to scrutinize the expression levels of MMP13, Collagen II, RUNX2, and aggrecan. Additionally, we conducted Co-IP assays on ZnCl2-treated ATDC5 cells to explore the interaction between SOX9 and SUMO1. Our investigation unveiled that zinc deficiency led to a reduction in cartilage development, as evidenced by the HE results in fetal murine femur. Moreover, diminished expression levels of SUMO1 and UBC9 were observed in the IHC and Western blot results. Furthermore, Western blot and Co-IP assays revealed an augmented interaction between SOX9 and SUMO1, which was potentiated by ZnCl2 treatment. Significantly, mutations at the SUMOylation site of SOX9 resulted in alterations in the expression patterns of crucial chondrogenesis factors. This research underscores how zinc ions promote cartilage development through the modification of SOX9 by SUMO1.
Collapse
Affiliation(s)
- Na Xue
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, China
- Emergency Medicine Department, Tianjin Fifth Central Hospital, Tianjin, China
| | - Jing Zhao
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Jing Yin
- Department of Pathology, Tianjin Fifth Central Hospital, Tianjin, China
| | - Liang Liu
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Zhong Yang
- Department of Orthopedics, Tianjin Fifth Central Hospital, Tianjin, China
| | - Shuchao Zhai
- Department of Orthopedics, Tianjin Fifth Central Hospital, Tianjin, China
| | - Xiyun Bian
- Tianjin Key Laboratory of Epigenetics for Organ Development of Preterm Infants, Central Laboratory, Tianjin Fifth Central Hospital, Tianjin, China
| | - Xiang Gao
- Department of Orthopedics, Tianjin Fifth Central Hospital, Tianjin, China
| |
Collapse
|
2
|
Shi J, He C, Chen L, Xing X, Wei W, Zhang J. Targeting PD-1 post-translational modifications for improving cancer immunotherapy. CELL INSIGHT 2025; 4:100248. [PMID: 40336591 PMCID: PMC12056969 DOI: 10.1016/j.cellin.2025.100248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/09/2025]
Abstract
Programmed cell death protein 1 (PD-1) is a critical immune checkpoint receptor that suppresses immune responses largely through its interaction with PD-L1. Tumors exploit this mechanism to evade immune surveillance, positioning immune checkpoint inhibitors targeting the PD-1/PD-L1 axis as groundbreaking advancements in cancer therapy. However, the overall effectiveness of these therapies is often constrained by an incomplete understanding of the underlying mechanisms. Recent research has uncovered the pivotal role of various post-translational modifications (PTMs) of PD-1, including ubiquitination, UFMylation, phosphorylation, palmitoylation, and glycosylation, in regulating its protein stability, localization, and protein-protein interactions. As much, dysregulation of these PTMs can drive PD-1-mediated immune evasion and contribute to therapeutic resistance. Notably, targeting PD-1 PTMs with small-molecule inhibitors or monoclonal antibodies (MAbs) has shown potential to bolster anti-tumor immunity in both pre-clinical mouse models and clinical trials. This review highlights recent findings on PD-1's PTMs and explores emerging therapeutic strategies aimed at modulating these modifications. By integrating these mechanistic insights, the development of combination cancer immunotherapies can be further rationally advanced, offering new avenues for more effective and durable treatments.
Collapse
Affiliation(s)
- Jie Shi
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Chuan He
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Li Chen
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Xixin Xing
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Jinfang Zhang
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Wuhan University, Wuhan 430072, Hubei, China
| |
Collapse
|
3
|
Yang W, Lefebvre V. PTPN11 in cartilage development, adult homeostasis, and diseases. Bone Res 2025; 13:53. [PMID: 40379623 DOI: 10.1038/s41413-025-00425-0] [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: 01/24/2025] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 05/19/2025] Open
Abstract
The SH2 domain-containing protein tyrosine phosphatase 2 (SHP2, also known as PTP2C), encoded by PTPN11, is ubiquitously expressed and has context-specific effects. It promotes RAS/MAPK signaling downstream of receptor tyrosine kinases, cytokine receptors, and extracellular matrix proteins, and was shown in various lineages to modulate cell survival, proliferation, differentiation, and migration. Over the past decade, PTPN11 inactivation in chondrocytes was found to cause metachondromatosis, a rare disorder characterized by multiple enchondromas and osteochondroma-like lesions. Moreover, SHP2 inhibition was found to mitigate osteoarthritis pathogenesis in mice, and abundant but incomplete evidence suggests that SHP2 is crucial for cartilage development and adult homeostasis, during which its expression and activity are tightly regulated transcriptionally and posttranslationally, and by varying sets of functional partners. Fully uncovering SHP2 actions and regulation in chondrocytes is thus fundamental to understanding the mechanisms underlying both rare and common cartilage diseases and to designing effective disease treatments. We here review current knowledge, highlight recent discoveries and controversies, and propose new research directions to answer remaining questions.
Collapse
Affiliation(s)
- Wentian Yang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI, USA.
| | - Véronique Lefebvre
- Division of Orthopaedic Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Wang Y, Ohnuki H, Tran AD, Wang D, Ha T, Feng JX, Sim M, Barnhill R, Lugassy C, Sargen MR, Salazar-Cavazos E, Kruhlak M, Tosato G. Induced clustering of SHP2-depleted tumor cells in vascular islands restores sensitivity to MEK/ERK inhibition. J Clin Invest 2025; 135:e181609. [PMID: 40131370 PMCID: PMC12077907 DOI: 10.1172/jci181609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Allosteric inhibitors of the tyrosine phosphatase Src homology 2 domain-containing protein tyrosine phosphatase 2 (SHP2) hold therapeutic promise in cancers with overactive RAS/ERK signaling, but adaptive resistance to SHP2 inhibitors may limit benefits. Here, we utilized tumor cells that proliferate similarly with or without endogenous SHP2 to explore means to overcome this growth independence from SHP2. We found that SHP2 depletion profoundly altered the output of vascular regulators, cytokines, chemokines, and other factors from SHP2 growth-resistant cancer cells. Tumors derived from inoculation of SHP2-depleted, but SHP2 growth-independent, mouse melanoma and colon carcinoma cell lines displayed a typically subverted architecture, in which proliferative tumor cells surrounding a remodeled vessel formed "vascular islands", each limited by surrounding hypoxic and dead tumor tissue, where inflammatory blood cells were limited. Although vascular islands generally reflect protected sanctuaries for tumor cells, we found that vascular island-resident, highly proliferative, SHP2-depleted tumor cells acquired an increased sensitivity to blockage of MEK/ERK signaling, resulting in reduced tumor growth. Our results show that the response to targeted therapies in resistant tumor cells was controlled by tumor cell-induced vascular changes and tumor architectural reorganization, providing a compelling approach to elicit tumor responses by exploiting tumor- and endothelium-dependent biochemical changes.
Collapse
MESH Headings
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency
- Animals
- Mice
- MAP Kinase Signaling System/drug effects
- Humans
- Cell Line, Tumor
- Drug Resistance, Neoplasm
- Colonic Neoplasms/pathology
- Colonic Neoplasms/drug therapy
- Melanoma, Experimental/pathology
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/enzymology
- Melanoma, Experimental/genetics
- Cell Proliferation
- Neovascularization, Pathologic/enzymology
Collapse
Affiliation(s)
- Yuyi Wang
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Hidetaka Ohnuki
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Andy D. Tran
- Center for Cancer Research Microscopy Core, Laboratory of Cancer Biology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Dunrui Wang
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Taekyu Ha
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Jing-Xin Feng
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Minji Sim
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Raymond Barnhill
- Department of Translational Research, Institut Curie, Paris, France
| | - Claire Lugassy
- Department of Translational Research, Institut Curie, Paris, France
| | - Michael R. Sargen
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, Maryland, USA
| | - Emanuel Salazar-Cavazos
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, NIH, Bethesda, Maryland, USA
| | - Michael Kruhlak
- Center for Cancer Research Microscopy Core, Laboratory of Cancer Biology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| |
Collapse
|
5
|
Rafiq H, Han L, Rehman AU, He P, Abdelhameed AS, Hassan ESG, Fu H, Wadood A, Hu J. Deciphering the structural and dynamic effects of SHP2-E76 mutations: mechanistic insights into oncogenic activation. BMC Chem 2025; 19:128. [PMID: 40369628 PMCID: PMC12079960 DOI: 10.1186/s13065-025-01494-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/30/2025] [Indexed: 05/16/2025] Open
Abstract
The tyrosine phosphatase known as SHP2 is a cytoplasmic protein and encodes by proto-oncogene PTPN11. This protein is essential for the regulation of cell growth, differentiation, programed cell death, and survival. This regulation is achieved through the release of intramolecular autoinhibition and the modulation of several signaling pathways, including the signaling cascade of Ras-MAPK. Mutations in SHP2 are frequently associated with human malignancies and neurodevelopmental disorders (NDDs). Specifically, a germline mutation (E76D) in SHP2 is linked to neurodevelopmental disorders, such as Noonan syndrome, while somatic mutations (E76G and E76A) and altered SHP2 expression are implicated in several forms of leukemia. These mutations disrupt the closed conformation, which normally keeps SHP2 in an inactive, auto-inhibited state, thereby enhancing phosphatase activity and activating SHP2, leading to a gain-of-function effect. However, the structural and functional implications of these disease-related mutants are not well elucidated. Therefore, in this study, we investigate the structural mechanisms underlying three distinct gain-of-function SHP2 mutations (E76D, E76G, and E76A) through the application of molecular dynamics (MD) simulations, focusing on how a single amino acid mutation at the same position result in different disease phenotypes, either cause cancer or NDDs. Notably, Patients with Noonan Syndrome have an increased risk of developing cancer, suggesting a potential link between these diseases and their mutations. MD simulation was employed to elucidate this mechanism, examining four distinct states: Apo-state (E76), M1-state (E76D), M2-state (E76G), and M3-state (E76A). The dynamics and conformational changes of SHP2 in both its Apo-state and mutant states (M1, M2, and M3) were compared. Our findings indicate that both cancer-related and NDD-related mutations destabilize the N-SH2 and PTP interface, facilitating SHP2 activation. However, the cancer-associated mutations induce more severe disruption at the N-SH2 and PTP interface than the NDD mutations. Additionally, dynamic analyses revealed that mutations at the interface (M1, M2, and M3) not only alter the native folded conformation of SHP2 but also significantly enhance the C-distance between the N-SH2 and PTP domains. Overall, this study provides a comprehensive understanding of the structural dynamics of SHP2 at the atomic level, revealing how mutations disrupt its auto-inhibition and increase PTP activity, providing valuable insights into the molecular mechanisms driving both cancer and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Humaira Rafiq
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan-23200, Pakistan
| | - Lu Han
- Department of Central Laboratory, SSL, Central Hospital of Dongguan City, Affiliated Dongguan Shilong People's Hospital of Guangdong Medical University, Dongguan, China
| | - Ashfaq Ur Rehman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, 92697-3900, USA
| | - Pei He
- The Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ali Saber Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, 11451, Riyadh, Saudi Arabia
| | - Eman S G Hassan
- Pharmacology Department, Egyptian Drug Authority (EDA), Formerly National Organization for Drug Control and Research (NODCAR), Cairo, Egypt
| | - Hongxia Fu
- Department of Neurology, SSL, Central Hospital of Dongguan City, Affiliated Dongguan Shilong People's Hospital of Guangdong Medical University, Dongguan, China
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan-23200, Pakistan.
| | - Junjian Hu
- Department of Central Laboratory, SSL, Central Hospital of Dongguan City, Affiliated Dongguan Shilong People's Hospital of Guangdong Medical University, Dongguan, China.
| |
Collapse
|
6
|
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
|
7
|
Levinson A, Shannon K, Huang BJ. Targeting Hyperactive Ras Signaling in Pediatric Cancer. Cold Spring Harb Perspect Med 2025; 15:a041572. [PMID: 39009442 PMCID: PMC12047744 DOI: 10.1101/cshperspect.a041572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Somatic RAS mutations are among the most frequent drivers in pediatric and adult cancers. Somatic KRAS, NRAS, and HRAS mutations exhibit distinct tissue-specific predilections. Germline NF1 and RAS mutations in children with neurofibromatosis type 1 and other RASopathy developmental disorders have provided new insights into Ras biology. In many cases, these germline mutations are associated with increased cancer risk. Promising targeted therapeutic strategies for pediatric cancers and neoplasms with NF1 or RAS mutations include inhibition of downstream Ras effector pathways, directly inhibiting the signal output of oncogenic Ras proteins and associated pathway members, and therapeutically targeting Ras posttranslational modifications and intracellular trafficking. Acquired drug resistance to targeted drugs remains a significant challenge but, increasingly, rational drug combination approaches have shown promise in overcoming resistance. Developing predictive preclinical models of childhood cancers for drug testing is a high priority for the field of pediatric oncology.
Collapse
Affiliation(s)
- Anya Levinson
- Department of Pediatrics, University of California San Francisco, San Francisco, California 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, USA
| | - Kevin Shannon
- Department of Pediatrics, University of California San Francisco, San Francisco, California 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, USA
| | - Benjamin J Huang
- Department of Pediatrics, University of California San Francisco, San Francisco, California 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
8
|
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:e2400938. [PMID: 40318117 DOI: 10.1002/cbic.202400938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
9
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
10
|
Doi T, Ishikawa T, Moriguchi M, Itoh Y. Current status of cancer genome medicine for pancreatic ductal adenocarcinoma. Jpn J Clin Oncol 2025; 55:443-452. [PMID: 39893577 DOI: 10.1093/jjco/hyaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis; however, advancements in cancer genome profiling using next-generation sequencing have provided new perspectives. KRAS mutations are the most frequently observed genomic alterations in patients with PDAC. However, until recently, it was not considered a viable therapeutic target. Although KRAS G12C mutations for which targeted therapies are already available are infrequent in PDAC, treatments targeting KRAS G12D and pan-KRAS are still under development. Similarly, new treatment methods for KRAS, such as chimeric antigen receptor T-cell therapy, have been developed. Several other potential therapeutic targets have been identified for KRAS wild-type PDAC. For instance, immune checkpoint inhibitors have demonstrated efficacy in PDAC treatment with microsatellite instability-high/deficient mismatch repair and tumor mutation burden-high profiles. However, for other PDAC cases with low immunogenicity, combination therapies that enhance the effectiveness of immune checkpoint inhibitors are being considered. Additionally, homologous recombination repair deficiencies, including BRCA1/2 mutations, are prevalent in PDAC and serve as important biomarkers for therapies involving poly (adenosine diphosphate-ribose) polymerase inhibitors and platinum-based therapies. Currently, olaparib is available for maintenance therapy of BRCA1/2 mutation-positive PDAC. Further therapeutic developments are ongoing for genetic abnormalities involving BRAF V600E and the fusion genes RET, NTRK, NRG, ALK, FGFR2, and ROS1. Overcoming advanced PDAC remains a formidable challenge; however, this review outlines the latest therapeutic strategies that are expected to lead to significant advancements.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Medical Oncology Unit, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
11
|
Lu J, Yu D, Li H, Qin P, Chen H, Chen L. Promising natural products targeting protein tyrosine phosphatase SHP2 for cancer therapy. Phytother Res 2025; 39:1735-1757. [PMID: 38558278 DOI: 10.1002/ptr.8185] [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: 09/01/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
The development of Src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2) inhibitors is a hot spot in the research and development of antitumor drugs, which may induce immunomodulatory effects in the tumor microenvironment and participate in anti-tumor immune responses. To date, several SHP2 inhibitors have made remarkable progress and entered clinical trials for the treatment of patients with advanced solid tumors. Multiple compounds derived from natural products have been proved to influence tumor cell proliferation, apoptosis, migration and other cellular functions, modulate cell cycle and immune cell activation by regulating the function of SHP2 and its mutants. However, there is a paucity of information about their diversity, biochemistry, and therapeutic potential of targeting SHP2 in tumors. This review will provide the structure, classification, inhibitory activities, experimental models, and antitumor effects of the natural products. Notably, this review summarizes recent advance in the efficacy and pharmacological mechanism of natural products targeting SHP2 in inhibiting the various signaling pathways that regulate different cancers and thus pave the way for further development of anticancer drugs targeting SHP2.
Collapse
Affiliation(s)
- Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danmei Yu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongtao Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pengcheng Qin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Henan University, Kaifeng, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Chen HZ, Kim NH, Nishizaki D, Nesline MK, Conroy JM, DePietro P, Pabla S, Kato S, Kurzrock R. PD-1 transcriptomic landscape across cancers and implications for immune checkpoint blockade outcome. NPJ Genom Med 2025; 10:21. [PMID: 40069238 PMCID: PMC11897377 DOI: 10.1038/s41525-025-00465-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/20/2025] [Indexed: 03/15/2025] Open
Abstract
Programmed cell death protein 1 (PD-1) is a critical immune checkpoint receptor and a target for cancer immune checkpoint inhibitors (ICI). We investigated PD-1 transcript expression across cancer types and its correlations to clinical outcomes. Using a reference population, PD-1 expression was calculated as percentiles in 489 of 514 patients (31 cancer types) with advanced/metastatic disease. PD-1 RNA expression varied across and within cancer types; pancreatic and liver/bile duct malignancies displayed the highest rates of high PD-1 (21.82% and 21.05%, respectively). Elevated CTLA-4, LAG-3, and TIGIT RNA expression were independently correlated with high PD-1. Although high PD-1 was not associated with outcome in immunotherapy-naïve patients (n = 272), in patients who received ICIs (n = 217), high PD-1 transcript expression was independently correlated with prolonged survival (hazard ratio 0.40; 95%CI, 0.18-0.92). This study identifies PD-1 as an important biomarker in predicting ICI outcomes, and advocates for comprehensive immunogenomic profiling in cancer management.
Collapse
Affiliation(s)
- Hui-Zi Chen
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Na Hyun Kim
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.
| | - Daisuke Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | | | | | | | | | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
13
|
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
|
14
|
Xu L, Jang H, Nussinov R. Allosteric modulation of NF1 GAP: Differential distributions of catalytically competent populations in loss-of-function and gain-of-function mutants. Protein Sci 2025; 34:e70042. [PMID: 39840811 PMCID: PMC11751910 DOI: 10.1002/pro.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/13/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
Neurofibromin (NF1), a Ras GTPase-activating protein (GAP), catalyzes Ras-mediated GTP hydrolysis and thereby negatively regulates the Ras/MAPK pathway. NF1 mutations can cause neurofibromatosis type 1 manifesting tumors, and neurodevelopmental disorders. Exactly how the missense mutations in the GAP-related domain of NF1 (NF1GRD) allosterically impact NF1 GAP to promote these distinct pathologies is unclear. Especially tantalizing is the question of how same-domain, same-residue NF1GRD variants exhibit distinct clinical phenotypes. Guided by clinical data, we take up this dilemma. We sampled the conformational ensembles of NF1GRD in complex with GTP-bound K-Ras4B by performing molecular dynamics simulations. Our results show that mutations in NF1GRD retain the active conformation of K-Ras4B but with biased propensities of the catalytically competent populations of K-Ras4B-NF1GRD complex. In agreement with clinical depiction and experimental tagging, compared to the wild type, NF1GRD E1356A and E1356V mutants effectively act through loss-of-function and gain-of-function mechanisms, leading to neurofibromatosis and developmental disorders, respectively. Allosteric modulation of NF1GRD GAP activity through biasing the conformational ensembles in the different states is further demonstrated by the diminished GAP activity by NF1GRD isoform 2, further manifesting propensities of conformational ensembles as powerful predictors of protein function. Taken together, our work identifies a NF1GRD hotspot that could allosterically tune GAP function, suggests targeting Ras oncogenic mutations by restoring NF1 catalytic activity, and offers a molecular mechanism for NF1 phenotypes determined by their distinct conformational propensities.
Collapse
Affiliation(s)
- Liang Xu
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation LaboratoryNational Cancer InstituteFrederickMarylandUSA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation LaboratoryNational Cancer InstituteFrederickMarylandUSA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation LaboratoryNational Cancer InstituteFrederickMarylandUSA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
15
|
Bennett AM, Tiganis T. Protein Tyrosine Phosphatases in Metabolism: A New Frontier for Therapeutics. Annu Rev Physiol 2025; 87:301-324. [PMID: 39531392 DOI: 10.1146/annurev-physiol-022724-105540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The increased prevalence of chronic metabolic disorders, including obesity and type 2 diabetes and their associated comorbidities, are among the world's greatest health and economic challenges. Metabolic homeostasis involves a complex interplay between hormones that act on different tissues to elicit changes in the storage and utilization of energy. Such processes are mediated by tyrosine phosphorylation-dependent signaling, which is coordinated by the opposing actions of protein tyrosine kinases and protein tyrosine phosphatases (PTPs). Perturbations in the functions of PTPs can be instrumental in the pathophysiology of metabolic diseases. The goal of this review is to highlight key advances in our understanding of how PTPs control body weight and glucose metabolism, as well as their contributions to obesity and type 2 diabetes. The emerging appreciation of the integrated functions of PTPs in metabolism, coupled with significant advances in pharmaceutical strategies aimed at targeting this class of enzymes, marks the advent of a new frontier in combating metabolic disorders.
Collapse
Affiliation(s)
- Anton M Bennett
- Yale Center for Molecular and Systems Metabolism, New Haven, Connecticut, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia;
| |
Collapse
|
16
|
Mitra R, Kumar S, Ayyannan SR. Identification of new small molecule allosteric SHP2 inhibitor through pharmacophore-based virtual screening, molecular docking, molecular dynamics simulation studies, synthesis and in vitro evaluation. J Biomol Struct Dyn 2025; 43:1352-1371. [PMID: 38095360 DOI: 10.1080/07391102.2023.2291733] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/15/2023] [Indexed: 01/16/2025]
Abstract
Src homology-2 (SH2) domain-containing phosphatase-2 (SHP2) is the first identified protooncogene and is a promising target for developing small molecule inhibitors as cancer chemotherapeutic agents. Pharmacophore-based virtual screening (PBVS) is a pharmacoinformatics methodology that employs physicochemical knowhow of the chemical space into the dynamic environs of computational technology to extract virtual molecular hits that are precise and promising for a drug target. In the current study, PBVS has been applied on EnamineTM Advanced Collection of 551,907 molecules by using a pharmacophore model developed upon SHP099 by Molecular Operating Environment (MOE) software to identify potential small molecule allosteric SHP2 inhibitors. Obtained 37 hits were further filtered through DruLiTo software for drug-likeness and PAINS remover which yielded 35 hits. These were subjected to molecular docking studies against the tunnel allosteric site of SHP2 (PDB ID: 5EHR) to screen them according to their binding affinity for the enzyme. Top 5 molecules having highest binding affinity for 5EHR were passed through an ADMET prediction screening and the top 2 hits (ligands 111675 and 546656) with the most favourable ADMET profile were taken for post screening molecular docking and MD simulation studies. From the protein-ligand interaction pattern, conformational stability and energy parameters, ligand 111675 (SHP2 Ki = 0.118 µM) resulted as the most active molecule. Further, the synthesis and in vitro evaluation of the lead compound 111675 unveiled its potent inhibitory activity (IC50 = 0.878 ± 0.008 µM) against SHP2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rangan Mitra
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi, Uttar Pradesh, India
| | - Sandeep Kumar
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi, Uttar Pradesh, India
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University) Varanasi, Uttar Pradesh, India
| |
Collapse
|
17
|
Makrides N, Sun E, Mir H, Jiang Z, Wu Y, Serra C, Cardoso WV, Shah NH, Zhang X. Allosteric inhibition rescues hydrocephalus caused by catalytically inactive Shp2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635289. [PMID: 39974929 PMCID: PMC11838390 DOI: 10.1101/2025.01.28.635289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
SHP2, a protein tyrosine phosphatase (PTP) crucial in Ras-MAPK signaling, is associated with various human congenital diseases and cancers. Here, we show that the catalytically inactive Shp2 C459S mutation results in communicating hydrocephalus, similar to the catalytically activating Shp2 E76K and Mek1 DD mutants. Unlike previous mutants, however, Shp2 C459S/+ mutation uniquely affects ciliary development rather than neurogenesis, leading to reduced cilia density and impaired ciliary motility. Differential scanning fluorimetry revealed that SHP2 C459S , SHP2 E76K and SHP2 C459S/E76K mutations all induce an open SHP2 conformation, but only SHP2 C459S leads to aberrant GAB1 phosphorylation in cells expressing wild-type SHP2. This distinctive signaling pattern correlates with our observations in brain ventricular tissues of Shp2 C459S/+ mice, where Erk and Stat3 activities remain normal but Gab1 phosphorylation is elevated. Critically, we show that the hydrocephalus phenotype in Shp2 C459S mice can be mitigated by allosteric inhibition of Shp2. These findings suggest that Shp2-associated hydrocephalus is driven by conformational changes rather than altered catalytic activity. Our results underscore the therapeutic potential of conformation-specific allosteric inhibitors in targeting both catalytically active and inactive SHP2 mutants.
Collapse
Affiliation(s)
- Neoklis Makrides
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Emily Sun
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Hilal Mir
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Ziyuan Jiang
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Yihua Wu
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Carlos Serra
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Wellington V Cardoso
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Neel H. Shah
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
18
|
Zhu C, Li L, Yu Y, Wang X, Shi Y, Gao Y, Chen K, Liu X, Cui Y, Zhang T, Yu Z. Optimization of SHP2 allosteric inhibitors with novel tail heterocycles and their potential as antitumor therapeutics. Eur J Med Chem 2025; 282:117078. [PMID: 39571459 DOI: 10.1016/j.ejmech.2024.117078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/10/2024]
Abstract
SHP2, a non-receptor protein tyrosine phosphatase involved in cancers, plays a pivotal role in numerous cellular signaling cascades, including the MAPK and PD-L1/PD-1 pathways. Although several SHP2 allosteric inhibitors have already entered clinical trials, none have been approved to date. Therefore, the development of new SHP2 allosteric inhibitors with improved efficacy is urgently required. Herein, we report the optimization of tail heterocycles in SHP2 allosteric inhibitors using a structure-based drug design strategy. Four series of compounds with different tail skeletons were synthesized, among which D13 showed notable inhibitory activity (IC50 = 1.2 μM) against SHP2. Molecular docking and binding studies indicated that the newly synthesized compounds exerted enzymatic inhibitory effects by directly binding to SHP2 with relatively slow dissociation rates. At the cellular level, Huh7 cells demonstrated heightened sensitivity to the novel SHP2 inhibitors, and D13 exhibited superior antiproliferative activity (IC50 = 38 μM) by arresting G0/G1 cell cycle, facilitating cell apoptosis and suppressing the MAPK signaling pathway. In the in vivo study, D13 displayed significant antitumor activity in a Huh7 xenograft model and possessed favorable druggability with acceptable oral bioavailability (F = 54 %) and half-life (t1/2 = 10.57 h). Collectively, this study lays a robust foundation for further optimization of the tail heterocycle skeleton in SHP2 allosteric inhibitors.
Collapse
Affiliation(s)
- Chengchun Zhu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Leilei Li
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Yan Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Xiao Wang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Ying Shi
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Yiping Gao
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Kai Chen
- Center for New Drug Evaluation, Shandong Academy of Pharmaceutical Sciences, Jinan, 250000, PR China
| | - Xiaoyu Liu
- Center for New Drug Evaluation, Shandong Academy of Pharmaceutical Sciences, Jinan, 250000, PR China
| | - Yuqian Cui
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 250012, PR China
| | - Tao Zhang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| | - Zhiyi Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
19
|
Pang J, Cen C, Tian Y, Cao X, Hao L, Tao X, Cao Z. Targeting Shp2 as a therapeutic strategy for neurodegenerative diseases. Transl Psychiatry 2025; 15:6. [PMID: 39794316 PMCID: PMC11724000 DOI: 10.1038/s41398-024-03222-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
The incidence of neurodegenerative diseases (NDs) has increased recently. However, most of the current governance strategies are palliative and lack effective therapeutic drugs. Therefore, elucidating the pathological mechanism of NDs is the key to the development of targeted drugs. As a member of the tyrosine phosphatase family, the role of Shp2 has been studied in tumors, but the research in the nervous system is still in a sporadic state. It can be phosphorylated by tyrosine kinases and then positively regulate tyrosine kinase-dependent signaling pathways. It could also be used as an adaptor protein to mediate downstream signaling pathways. Most of the existing studies have shown that Shp2 may be a potential molecular "checkpoint" against NDs, but its role in promoting degenerative lesions is difficult to ignore as well, and its two-way effect of both activation and inhibition is very distinctive. Shp2 is closely related to NDs-related pathogenic factors such as oxidative stress, mitochondrial dysfunction, excitatory toxicity, immune inflammation, apoptosis, and autophagy. Its bidirectional effects interfere with these pathogenic factors, making it a core component of the feedback and crosstalk network between multiple signaling pathways. Therefore, this article reviews the molecular mechanism of Shp2 regulation in NDs and its regulatory role in various pathogenic factors, providing evidence for the treatment of NDs by targeting Shp2 and the development of molecular targeted drugs.
Collapse
Affiliation(s)
- Jiao Pang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, PR China
- Department of Pathology and pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China
- College of Life Science, Northwest University, Xi'an City, Shaanxi Province, PR China
| | - Changqian Cen
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, PR China
| | - Yuan Tian
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Xingrui Cao
- Department of Chemistry, School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, PR China
| | - Liang Hao
- Department of Chemistry, School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, PR China.
| | - Xueshu Tao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, PR China.
- Department of Pain Medicine, The First Hospital of China Medical University, Shenyang, Liaoning Province, PR China.
| | - Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
20
|
Jeon S, Jeon Y, Lim JY, Kim Y, Cha B, Kim W. Emerging regulatory mechanisms and functions of biomolecular condensates: implications for therapeutic targets. Signal Transduct Target Ther 2025; 10:4. [PMID: 39757214 DOI: 10.1038/s41392-024-02070-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/01/2024] [Accepted: 11/06/2024] [Indexed: 01/07/2025] Open
Abstract
Cells orchestrate their processes through complex interactions, precisely organizing biomolecules in space and time. Recent discoveries have highlighted the crucial role of biomolecular condensates-membrane-less assemblies formed through the condensation of proteins, nucleic acids, and other molecules-in driving efficient and dynamic cellular processes. These condensates are integral to various physiological functions, such as gene expression and intracellular signal transduction, enabling rapid and finely tuned cellular responses. Their ability to regulate cellular signaling pathways is particularly significant, as it requires a careful balance between flexibility and precision. Disruption of this balance can lead to pathological conditions, including neurodegenerative diseases, cancer, and viral infections. Consequently, biomolecular condensates have emerged as promising therapeutic targets, with the potential to offer novel approaches to disease treatment. In this review, we present the recent insights into the regulatory mechanisms by which biomolecular condensates influence intracellular signaling pathways, their roles in health and disease, and potential strategies for modulating condensate dynamics as a therapeutic approach. Understanding these emerging principles may provide valuable directions for developing effective treatments targeting the aberrant behavior of biomolecular condensates in various diseases.
Collapse
Affiliation(s)
- Soyoung Jeon
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Yeram Jeon
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Ji-Youn Lim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Yujeong Kim
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Boksik Cha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea.
| | - Wantae Kim
- Department of Life Science, University of Seoul, Seoul, South Korea.
| |
Collapse
|
21
|
Wang K, Zhang X, Hu Y, Guo J, Shen G, Zhang K, Jiang S, Wang T. Discovery of novel phenyl urea SHP2 inhibitors with anti-colon cancer and potential immunomodulatory effects. Eur J Med Chem 2025; 281:117036. [PMID: 39541871 DOI: 10.1016/j.ejmech.2024.117036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Src Homology-2 Domain Containing Protein Tyrosine Phosphatase-2 (SHP2) is a non-receptor-type protein tyrosine phosphatase (PTP), which is recognized as potential and attractive cancer therapeutic target. Currently, no SHP2 inhibitors have been approved for clinical use, and colorectal cancer (CRC) cells exhibited frequent resistance to reported SHP2 inhibitors, such as SHP099 and TNO155. Herein, we reported our discovery and optimization of phenyl urea as novel SHP2 inhibitors. A8, the most potential SHP2 inhibitor, exhibited great antiproliferative activities against SHP099/TNO155-insensitive tumor cell lines, and rescued PD-L1-mediated immunosuppression. A8 significantly suppressed in vivo tumor growth in a CT26 mouse model and activated immunomodulatory effects in tumor microenvironment. Our work demonstrated that A8 has the potential to be a lead compound for the further development of SHP2 inhibitor and the treatment of CRC.
Collapse
Affiliation(s)
- Kaizhen Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiangyu Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yingxin Hu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiazheng Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Guoqing Shen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kuojun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Sheng Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
22
|
Sasmal P, Prabitha P, Prashantha Kumar BR, Swetha BR, Babasahib SK, Raghavendra NM. Beyond peptides: Unveiling the design strategies, structure activity correlations and protein-ligand interactions of small molecule inhibitors against PD-1/PD-L1. Bioorg Chem 2025; 154:108036. [PMID: 39693923 DOI: 10.1016/j.bioorg.2024.108036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
The landscape of cancer treatment has been transformed by the emergence of immunotherapy, especially through the use of antibodies that target the PD-1/PD-L1 pathway. Recently, there has been a notable increase in interest surrounding immune checkpoint inhibitors for cancer therapy. While antibody-based approaches have drawbacks like high costs and prolonged activity, the approval of monoclonal antibodies such as pembrolizumab and nivolumab has paved the way for a range of alternative therapies, including peptides, peptidomimetics, and small-molecule inhibitors. These smaller molecules, which target the PD-1/PD-L1 interaction, are seen as potential substitutes or supplements to monoclonal antibodies. Our focus in this article is primarily on exploring small molecules designed for PD-1/PD-L1 checkpoint pathway modulation in cancer immunotherapy, along with highlighting current advances in their structural and preclinical/clinical development. The pursuit of therapeutics based on small-molecule inhibitors of the PD-1/PD-L1 axis offers a promising yet intricate avenue for advancing cancer treatment.
Collapse
Affiliation(s)
- Pujan Sasmal
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar - 160 062, Punjab, India; Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy (ABMRCP), Bengaluru 560 107, Karnataka, India.
| | - P Prabitha
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - B R Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India
| | - B R Swetha
- Department of Pharmacology, College of Pharmaceutical Sciences, Dayananda Sagar University (DSU), Bengaluru 560 111, Karnataka, India
| | - Sajeev Kumar Babasahib
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University (DSU), Bengaluru 560 111, Karnataka, India
| | - Nulgumnalli Manjunathaiah Raghavendra
- Department of Pharmaceutical Chemistry, College of Pharmaceutical Sciences, Dayananda Sagar University (DSU), Bengaluru 560 111, Karnataka, India; Department of Pharmaceutical Chemistry, R R College of Pharmacy, Bengaluru 560 090, Karnataka, India.
| |
Collapse
|
23
|
Hammi I, Giron-Michel J, Akarid K, Arnoult D. FcRγIIA response duality in leishmaniasis. Microb Pathog 2025; 198:107123. [PMID: 39557223 DOI: 10.1016/j.micpath.2024.107123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/25/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
Leishmania is responsible for a neglected tropical disease affecting millions of people around the world and could potentially spread more due to climate change. This disease not only leads to significant morbidity but also imposes substantial social and economic burdens on affected populations, often exacerbating poverty and health disparities. Despite the complexity and effectiveness of the immune response, the parasite has developed various strategies to evade detection and manipulates host cells in favor of its replication. These evasion strategies start at early stages of the infection by hijacking immune receptors to silence critical cellular response that would otherwise limit the pathogen's propagation. Among these receptors, Fc receptors have emerged as a significant player in the immune evasion strategies employed by microorganisms, as they could promote inhibitory pathways. This review explores the potential role of one of these immune receptors, the FcγRIIA, in leishmaniasis and how this parasite may use it and the signaling pathways downstream to evade the host immune response. By understanding the potential interactions between Leishmania and immune receptors such as FcγRIIA, we may identify novel targets for therapeutic intervention aimed to enhance the host immune response and reduce the burden of this disease.
Collapse
Affiliation(s)
- Ikram Hammi
- Health & Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Morocco; INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France.
| | - Julien Giron-Michel
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France
| | - Khadija Akarid
- Health & Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Morocco
| | - Damien Arnoult
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France.
| |
Collapse
|
24
|
Xi R, Cao Y, Fu N, Sheng Y, Yu J, Li L, Zhang G, Wang F. Allosteric inhibition of the tyrosine phosphatase SHP2 enhances the anti-tumor immunity of interferon α through induction of caspase-1-mediated pyroptosis in renal cancer. Int Immunopharmacol 2024; 143:113498. [PMID: 39467353 DOI: 10.1016/j.intimp.2024.113498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Interferon alpha (IFNα) leads to therapeutic effects on various tumors, especially renal cell cancer (RCC), by directly protecting against tumors cell proliferation or indirectly inducing an anti-tumor immune response. However, new combination therapies are needed to enhance the efficacy of IFNα and reduce its adverse effects during long-term treatment. In this study, we found that the anti-proliferative effects of IFNα on RCC cells in vitro and in vivo were greater after the allosteric inhibition of SHP2 by SHP099 than after treatment with enzymatic inhibitors of SHP2. SHP099 increased IFNα-induced pro-caspase-1 expression in RCC cells, activated the NLRP3 inflammasome, and induced pyroptosis. Mechanistically, SHP099 not only increased the expression of NLRP3 inflammasome components via the NF-κB signaling pathway, but also further activated the NLRP3 inflammasome by regulating mitochondrial homeostasis through ANT1-mediated reactive oxygen species modulation. Allosteric inhibition of SHP2 by SHP099 also potently enhanced the anti-tumor immunity induced by IFNα by modulating T cell proliferation and infiltration in vitro and in vivo. These results reveal the new function of SHP2 in NLRP3 inflammasome activation and pyroptosis in RCC and provide a basis for further investigating the combination of allosteric SHP2 inhibitors with IFNα in cancer immunotherapy.
Collapse
Affiliation(s)
- Ruiying Xi
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Cao
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Naijie Fu
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuwen Sheng
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
| | - Jialing Yu
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingyu Li
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guolin Zhang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
| | - Fei Wang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China.
| |
Collapse
|
25
|
Hammi I, Giron-Michel J, Riyad M, Akarid K, Arnoult D. FcRγIIA attenuates pathology of cutaneous leishmaniasis and modulates ITAMa/i balance. Parasit Vectors 2024; 17:517. [PMID: 39696675 DOI: 10.1186/s13071-024-06593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Leishmania is the causal parasite of leishmaniasis, a neglected tropical disease affecting millions of individuals worldwide, and its dissemination is linked to climate change. Despite the complexity and effectiveness of the immune response, the parasite has developed many strategies to evade it and take control of the host cell to replicate. These evasion strategies start at early stages of infection by hijacking immune receptors to mitigate the cellular response. In this study, we examined whether Leishmania uses the Fc receptor FcγRIIA/CD32a and its downstream signaling pathways to evade the host immune response. METHODS Regarding in vivo studies, CD32a transgenic mice and the corresponding wild types were infected with Leishmania major Friedlin strain. For the in vitro experiments, BMDMs isolated from WT or CD32a transgenic mice and control or CD32a knockdown differentiated THP-1s were infected with two species of Leishmania, Leishmania major and L. tropica. RESULTS In vivo, expression of FcγRIIA/CD32a was found to accelerate the signs of inflammation while simultaneously preventing the formation of necrotic lesions after Leishmania infection. In infected macrophages, the presence of FcγRIIA/CD32a did not affect the secretion of proinflammatory cytokines, while the balance between ITAMa and ITAMi proteins was disturbed with improved Fyn and Lyn activation. Unexpectedly, infection with L. tropica but not L. major triggered an intracytoplasmic processing of FcγRIIA/CD32a. CONCLUSIONS Our observations underscore the significance of FcγRIIA/CD32a in cutaneous leishmaniasis and its potential use as a therapeutic target.
Collapse
Affiliation(s)
- Ikram Hammi
- Health & Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca, Morocco
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France
| | - Julien Giron-Michel
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France
| | - Myriam Riyad
- Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, UH2C, Casablanca, Morocco
| | - Khadija Akarid
- Health & Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca, Morocco
| | - Damien Arnoult
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France.
| |
Collapse
|
26
|
Chen J, Li W, Zhang C, Wen D, Jiao C. Tyrosine phosphatase SHP2 promoted the progression of CRC via modulating the PI3K/BRD4/TFEB signaling induced ferroptosis. Discov Oncol 2024; 15:793. [PMID: 39692787 DOI: 10.1007/s12672-024-01586-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/13/2024] [Indexed: 12/19/2024] Open
Abstract
OBJECTIVE To elucidate the mechanism by which tyrosine phosphatase SHP2 protects CRC through modulation of TFEB-mediated ferritinophagy, thereby suppressing ROS and ferroptosis. METHODS SW480 and SW620 cells, in the logarithmic growth phase, were treated with or without the SHP2 inhibitor PHPS1, the activator Trichomide A, EGF, or MMP inhibitors, and randomly assigned to four groups. Additionally, SW480 cells in the logarithmic phase underwent treatments with EGF, the ferroptosis inducer erastin, Trichomide A, or the curcumin analog C1, forming seven groups. Cell migration assessment in these groups employed scratch and Transwell assays. Protein expression analysis of total SHP2, total PI3K, p-SHP2, p-PI3K, p-TFEB, TFEB, SQSTM1, LC3, LAMP2, NCOA4, FTH1, GPX4, NOX4, and ACSL4 in the seven SW480 groups was conducted through Western blot and immunofluorescence. Apoptosis analysis was performed on these seven groups, while gene co-expression analysis utilized bioinformatics. SW480 and CCD-841CoN cells were categorized into four groups, undergoing treatment with saline, EGFR-OE lentivirus, SHP2-KD lentivirus, or SHP2-OE lentivirus. Western blot analysis in SW480 cells detected EGFR, total SHP2, p-SHP2, GPX4, and ACSL4 proteins, and tumor volume observations were conducted in a nude mouse xenograft model. Western blot also evaluated total SHP2, p-SHP2, GPX4, and ACSL4 protein expression in CCD-841CoN cells. RESULTS Bioinformatics analysis revealed correlations between EGFR and SHP2, SHP2 and PIK3CA, SHP2 and MAPK1, BRK4 and HIF1A, HIF1A and NCOA4, as well as TFEB and FTH1. Scratch and Transwell assays showed that SHP2 diminishes the migratory capacity of SW480 and SW620 cells. Western blot and immunofluorescence demonstrated that EGFR activation of SHP2 markedly elevated p-TFEB levels while reducing TFEB protein expression. EGF stimulation enhanced the expression of FTH1, GPX4, NOX4, and ACSL4. Combined stimulation with EGF and SHP2 further amplified the expression of p-SHP2, p-TFEB, and NCOA4 while reducing TFEB, SQSTM1, LC3, and LAMP2. Erastin augmented FTH1, GPX4, NOX4, and ACSL4 expression while decreasing p-SHP2, p-TFEB, TFEB, SQSTM1, LC3, LAMP2, and NCOA4. TFEB activation suppressed p-SHP2, p-TFEB, NCOA4, FTH1, and GPX4 expression, while promoting TFEB, SQSTM1, LC3, LAMP2, NOX4, and ACSL4 expression. Apoptosis assays indicated that SHP2 activation decelerated apoptosis in SW480 cells, whereas erastin under EGF stimulation accelerated apoptosis, as did TFEB activation. Western blot results in SW480 cells displayed that overexpression of EGFR or SHP2 significantly increased total SHP2, p-SHP2, and GPX4 expression while decreasing ACSL4 levels. SHP2 knockdown decreased total SHP2, p-SHP2, and GPX4 expression, with an increase in ACSL4 expression. In CCD-841CoN cells, overexpression of EGFR or SHP2 resulted in a decrease in p-SHP2 and an increase in total SHP2, more pronounced with SHP2 overexpression, while GPX4 and ACSL4 levels remained stable. SHP2 knockdown led to reduced EGFR, total SHP2, p-SHP2, and GPX4 expression, without a significant impact on ACSL4 levels. The nude mouse xenograft model demonstrated that EGFR overexpression significantly increased tumor size, whereas SHP2 overexpression markedly decreased tumor volume. SHP2 knockdown resulted in significantly larger tumors. CONCLUSION SHP2 advances CRC progression by modulating TFEB-mediated ferritinophagy, suppressing ROS and ferroptosis. Targeting SHP2 presents a promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Jian Chen
- Department of General Surgery, Bethune International Peace Hospital of The People's Liberation Army, No. 398, Zhongshan XI Road, Qiaoxi District, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Wei Li
- Department of General Surgery, Bethune International Peace Hospital of The People's Liberation Army, No. 398, Zhongshan XI Road, Qiaoxi District, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Cheng Zhang
- Department of Gastroenterology, Bethune International Peace Hospital of The People's Liberation Army, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Dihao Wen
- Department of General Surgery, Bethune International Peace Hospital of The People's Liberation Army, No. 398, Zhongshan XI Road, Qiaoxi District, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Cheng Jiao
- Department of General Surgery, Bethune International Peace Hospital of The People's Liberation Army, No. 398, Zhongshan XI Road, Qiaoxi District, Shijiazhuang, 050000, Hebei, People's Republic of China.
| |
Collapse
|
27
|
Sampson J, Ju HM, Zhang N, Yeoh S, Choi J, Bayliss R. Targeting ERBB3 and AKT to overcome adaptive resistance in EML4-ALK-driven non-small cell lung cancer. Cell Death Dis 2024; 15:912. [PMID: 39695132 DOI: 10.1038/s41419-024-07272-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
The fusion event between EML4 and ALK drives a significant oncogenic activity in 5% of non-small cell lung cancer (NSCLC). Even though potent ALK-tyrosine kinase inhibitors (ALK-TKIs) are successfully used for the treatment of EML4-ALK-positive NSCLC patients, a subset of those patients eventually acquire resistance during their therapy. Here, we investigate the kinase responses in EML4-ALK V1 and V3-harbouring NSCLC cancer cells after acute inhibition with ALK TKI, lorlatinib (LOR). Using phosphopeptide chip array and upstream kinase prediction analysis, we identified a group of phosphorylated tyrosine peptides including ERBB and AKT proteins that are upregulated upon ALK-TKI treatment in EML4-ALK-positive NSCLC cell lines. Dual inhibition of ALK and ERBB receptors or AKT disrupts RAS/MAPK and AKT/PI3K signalling pathways, and enhances apoptosis in EML4-ALK + NSCLC cancer cells. Heregulin, an ERBB3 ligand, differentially modulates the sensitivity of EML4-ALK cell lines to ALK inhibitors. We found that EML4-ALK cells made resistant to LOR are sensitive to inhibition of ERBB and AKT. These findings emphasize the important roles of AKT and ERBB3 to regulate signalling after acute LOR treatment, identifying them as potential targets that may be beneficial to prevent adaptive resistance to EML4-ALK-targeted therapies in NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Receptor, ErbB-3/metabolism
- Receptor, ErbB-3/genetics
- Oncogene Proteins, Fusion/metabolism
- Oncogene Proteins, Fusion/genetics
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Lactams/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Aminopyridines/pharmacology
- Signal Transduction/drug effects
- Pyrazoles/pharmacology
- Neuregulin-1/metabolism
- Neuregulin-1/genetics
- Lactams, Macrocyclic/pharmacology
- Anaplastic Lymphoma Kinase/genetics
- Anaplastic Lymphoma Kinase/metabolism
- Anaplastic Lymphoma Kinase/antagonists & inhibitors
- Apoptosis/drug effects
- ErbB Receptors/metabolism
- ErbB Receptors/genetics
- ErbB Receptors/antagonists & inhibitors
Collapse
Affiliation(s)
- Josephina Sampson
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| | - Hyun-Min Ju
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Korea
| | - Nan Zhang
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Sharon Yeoh
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Jene Choi
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Korea.
| | - Richard Bayliss
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
28
|
Pham L, Arroum T, Wan J, Pavelich L, Bell J, Morse PT, Lee I, Grossman LI, Sanderson TH, Malek MH, Hüttemann M. Regulation of mitochondrial oxidative phosphorylation through tight control of cytochrome c oxidase in health and disease - Implications for ischemia/reperfusion injury, inflammatory diseases, diabetes, and cancer. Redox Biol 2024; 78:103426. [PMID: 39566165 PMCID: PMC11617887 DOI: 10.1016/j.redox.2024.103426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024] Open
Abstract
Mitochondria are essential to cellular function as they generate the majority of cellular ATP, mediated through oxidative phosphorylation, which couples proton pumping of the electron transport chain (ETC) to ATP production. The ETC generates an electrochemical gradient, known as the proton motive force, consisting of the mitochondrial membrane potential (ΔΨm, the major component in mammals) and ΔpH across the inner mitochondrial membrane. Both ATP production and reactive oxygen species (ROS) are linked to ΔΨm, and it has been shown that an imbalance in ΔΨm beyond the physiological optimal intermediate range results in excessive ROS production. The reaction of cytochrome c oxidase (COX) of the ETC with its small electron donor cytochrome c (Cytc) is the proposed rate-limiting step in mammals under physiological conditions. The rate at which this redox reaction occurs controls ΔΨm and thus ATP and ROS production. Multiple mechanisms are in place that regulate this reaction to meet the cell's energy demand and respond to acute stress. COX and Cytc have been shown to be regulated by all three main mechanisms, which we discuss in detail: allosteric regulation, tissue-specific isoforms, and post-translational modifications for which we provide a comprehensive catalog and discussion of their functional role with 55 and 50 identified phosphorylation and acetylation sites on COX, respectively. Disruption of these regulatory mechanisms has been found in several common human diseases, including stroke and myocardial infarction, inflammation including sepsis, and diabetes, where changes in COX or Cytc phosphorylation lead to mitochondrial dysfunction contributing to disease pathophysiology. Identification and subsequent targeting of the underlying signaling pathways holds clear promise for future interventions to improve human health. An example intervention is the recently discovered noninvasive COX-inhibitory infrared light therapy that holds promise to transform the current standard of clinical care in disease conditions where COX regulation has gone awry.
Collapse
Affiliation(s)
- Lucynda Pham
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA.
| | - Tasnim Arroum
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA.
| | - Junmei Wan
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA.
| | - Lauren Pavelich
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI, 48201, USA.
| | - Jamie Bell
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA; Division of Pediatric Critical Care, Children's Hospital of Michigan, Central Michigan University, Detroit, MI, 48201, USA.
| | - Paul T Morse
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA.
| | - Icksoo Lee
- College of Medicine, Dankook University, Cheonan-si, 31116, Republic of Korea.
| | - Lawrence I Grossman
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA.
| | - Thomas H Sanderson
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| | - Moh H Malek
- Department of Health Care Sciences, Eugene Applebaum College of Pharmacy & Health Sciences, Wayne State University, Detroit, MI, 48201, USA.
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University, Detroit, MI, 48201, USA.
| |
Collapse
|
29
|
Marasco M, Kirkpatrick J, Carlomagno T, Hub JS, Anselmi M. Phosphopeptide binding to the N-SH2 domain of tyrosine phosphatase SHP2 correlates with the unzipping of its central β-sheet. Comput Struct Biotechnol J 2024; 23:1169-1180. [PMID: 38510972 PMCID: PMC10951427 DOI: 10.1016/j.csbj.2024.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
SHP2 is a tyrosine phosphatase that plays a regulatory role in multiple intracellular signaling cascades and is known to be oncogenic in certain contexts. In the absence of effectors, SHP2 adopts an autoinhibited conformation with its N-SH2 domain blocking the active site. Given the key role of N-SH2 in regulating SHP2, this domain has been extensively studied, often by X-ray crystallography. Using a combination of structural analyses and molecular dynamics (MD) simulations we show that the crystallographic environment can significantly influence the structure of the isolated N-SH2 domain, resulting in misleading interpretations. As an orthogonal method to X-ray crystallography, we use a combination of NMR spectroscopy and MD simulations to accurately determine the conformation of apo N-SH2 in solution. In contrast to earlier reports based on crystallographic data, our results indicate that apo N-SH2 in solution primarily adopts a conformation with a fully zipped central β-sheet, and that partial unzipping of this β-sheet is promoted by binding of either phosphopeptides or even phosphate/sulfate ions.
Collapse
Affiliation(s)
- Michelangelo Marasco
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John Kirkpatrick
- School of Biosciences, University of Birmingham, Edgbaston, B15 2TT Birmingham, UK
| | - Teresa Carlomagno
- School of Biosciences, University of Birmingham, Edgbaston, B15 2TT Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT Birmingham, UK
| | - Jochen S. Hub
- Theoretical Physics and Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| | - Massimiliano Anselmi
- Theoretical Physics and Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
30
|
Sun J, Lei D. CD200-CD200R Pathway: A Regulator of Microglial Polarization in Postoperative Cognitive Dysfunction. J Inflamm Res 2024; 17:8421-8427. [PMID: 39530002 PMCID: PMC11552426 DOI: 10.2147/jir.s489895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Microglial polarization refers to the ability of microglia to exhibit different functional states under various conditions. As the resident immune cells of the brain, changes in the functional state of microglia play a crucial role in the progression of postoperative cognitive dysfunction. Recent studies have indicated that CD200-CD200R signaling is associated with microglial polarization. This review focuses on the latest advancements regarding whether CD200-CD200R signaling can regulate microglial polarization and thereby influence postoperative cognitive dysfunction.
Collapse
Affiliation(s)
- Jie Sun
- Department of Anesthesiology, Zhongda Hospital Southeast University (Jiangbei), Nanjing, Jiangsu, 210044, People’s Republic of China
- Department of Anesthesiology, Zhongda Hospital Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| | - Daoyun Lei
- Department of Anesthesiology, Zhongda Hospital Southeast University (Jiangbei), Nanjing, Jiangsu, 210044, People’s Republic of China
- Department of Anesthesiology, Zhongda Hospital Southeast University, Nanjing, Jiangsu, 210009, People’s Republic of China
| |
Collapse
|
31
|
Gonzalez-Martinez D, Roth L, Mumford TR, Guan J, Le A, Doebele RC, Huang B, Tulpule A, Niewiadomska-Bugaj M, Bivona TG, Bugaj LJ. Oncogenic EML4-ALK assemblies suppress growth factor perception and modulate drug tolerance. Nat Commun 2024; 15:9473. [PMID: 39488530 PMCID: PMC11531495 DOI: 10.1038/s41467-024-53451-7] [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: 07/06/2024] [Accepted: 10/12/2024] [Indexed: 11/04/2024] Open
Abstract
Drug resistance remains a challenge for targeted therapy of cancers driven by EML4-ALK and related fusion oncogenes. EML4-ALK forms cytoplasmic protein condensates, which result from networks of interactions between oncogene and adapter protein multimers. While these assemblies are associated with oncogenic signaling, their role in drug response is unclear. Here, we use optogenetics and live-cell imaging to find that EML4-ALK assemblies suppress transmembrane receptor tyrosine kinase (RTK) signaling by sequestering RTK adapter proteins including GRB2 and SOS1. Furthermore, ALK inhibition, while suppressing oncogenic signaling, simultaneously releases the sequestered adapters and thereby resensitizes RTK signaling. Resensitized RTKs promote rapid and pulsatile ERK reactivation that originates from paracrine ligands shed by dying cells. Reactivated ERK signaling promotes cell survival, which can be counteracted by combination therapies that block paracrine signaling. Our results identify a regulatory role for RTK fusion assemblies and uncover a mechanism of tolerance to targeted therapies.
Collapse
Affiliation(s)
| | - Lee Roth
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Thomas R Mumford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Juan Guan
- Department of Physics, Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, 32611, USA
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, 32611, USA
| | - Anh Le
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Robert C Doebele
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, 94143, USA
- Department of Biochemistry and Biophysics, UCSF, San Francisco, 94143, USA
- Chan Zuckerberg Biohub, San Francisco, 94158, USA
| | - Asmin Tulpule
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | - Trever G Bivona
- Department of Medicine, Division of Hematology and Oncology, UCSF, San Francisco, CA, 94143, USA
| | - Lukasz J Bugaj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
32
|
Abbasi M, Gupta V, Chitranshi N, Moustardas P, Ranjbaran R, Graham SL. Molecular Mechanisms of Glaucoma Pathogenesis with Implications to Caveolin Adaptor Protein and Caveolin-Shp2 Axis. Aging Dis 2024; 15:2051-2068. [PMID: 37962455 PMCID: PMC11346403 DOI: 10.14336/ad.2023.1012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023] Open
Abstract
Glaucoma is a common retinal disorder characterized by progressive optic nerve damage, resulting in visual impairment and potential blindness. Elevated intraocular pressure (IOP) is a major risk factor, but some patients still experience disease progression despite IOP-lowering treatments. Genome-wide association studies have linked variations in the Caveolin1/2 (CAV-1/2) gene loci to glaucoma risk. Cav-1, a key protein in caveolae membrane invaginations, is involved in signaling pathways and its absence impairs retinal function. Recent research suggests that Cav-1 is implicated in modulating the BDNF/TrkB signaling pathway in retinal ganglion cells, which plays a critical role in retinal ganglion cell (RGC) health and protection against apoptosis. Understanding the interplay between these proteins could shed light on glaucoma pathogenesis and provide potential therapeutic targets.
Collapse
Affiliation(s)
- Mojdeh Abbasi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia.
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping Sweden.
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia.
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia.
| | - Petros Moustardas
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping Sweden.
| | - Reza Ranjbaran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Stuart L. Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia.
| |
Collapse
|
33
|
Gencer Akçok EB, Güner H, Akçok İ. Determination of promising inhibitors for N-SH2 domain of SHP2 tyrosine phosphatase: an in silico study. Mol Divers 2024; 28:3393-3407. [PMID: 38739228 PMCID: PMC11612003 DOI: 10.1007/s11030-024-10880-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/15/2024] [Indexed: 05/14/2024]
Abstract
There are many genes that produce proteins related to diseases and these proteins can be targeted with drugs as a potential therapeutic approach. Recent advancement in drug discovery techniques have created new opportunities for treating variety of diseases by targeting disease-related proteins. Structure-based drug discovery is a faster and more cost-effective approach than traditional methods. SHP2 phosphatase, encoded by the PTPN11 gene, has been the focus of much attention due to its involvement in many types of diseases. The biological function of SHP2 is enabled mostly by protein-protein interaction through its SH2 domains. In this study, we report the identification of a potential small molecule inhibitor for the N-SH2 domain of SHP2 by structure-based drug discovery approach. We utilized molecular docking studies, followed by molecular dynamics simulations and MM/PBSA calculations, to analyze compounds retrieved from the Broad's Drug Repurposing Hub and ZINC15 databases. We selected 10 hit compounds with the best docking scores from the libraries and examined their binding properties in the N-SH2 domain. We found that compound CID 60838 (Irinotecan) was the most suitable compound with a binding free energy value of - 64.45 kcal/mol and significant interactions with the target residues in the domain.
Collapse
Affiliation(s)
- Emel Başak Gencer Akçok
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gül University, 38080, Kayseri, Türkiye
| | - Hüseyin Güner
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gül University, 38080, Kayseri, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340, Balçova, İzmir, Türkiye
- Izmir Biomedicine and Genome Center (IBG), 35340, Balçova, İzmir, Türkiye
| | - İsmail Akçok
- Department of Bioengineering, Faculty of Life and Natural Sciences, Abdullah Gül University, 38080, Kayseri, Türkiye.
| |
Collapse
|
34
|
Hossain MA. Targeting the RAS upstream and downstream signaling pathway for cancer treatment. Eur J Pharmacol 2024; 979:176727. [PMID: 38866361 DOI: 10.1016/j.ejphar.2024.176727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Cancer often involves the overactivation of RAS/RAF/MEK/ERK (MAPK) and PI3K-Akt-mTOR pathways due to mutations in genes like RAS, RAF, PTEN, and PIK3CA. Various strategies are employed to address the overactivation of these pathways, among which targeted therapy emerges as a promising approach. Directly targeting specific proteins, leads to encouraging results in cancer treatment. For instance, RTK inhibitors such as imatinib and afatinib selectively target these receptors, hindering ligand binding and reducing signaling initiation. These inhibitors have shown potent efficacy against Non-Small Cell Lung Cancer. Other inhibitors, like lonafarnib targeting Farnesyltransferase and GGTI 2418 targeting geranylgeranyl Transferase, disrupt post-translational modifications of proteins. Additionally, inhibition of proteins like SOS, SH2 domain, and Ras demonstrate promising anti-tumor activity both in vivo and in vitro. Targeting downstream components with RAF inhibitors such as vemurafenib, dabrafenib, and sorafenib, along with MEK inhibitors like trametinib and binimetinib, has shown promising outcomes in treating cancers with BRAF-V600E mutations, including myeloma, colorectal, and thyroid cancers. Furthermore, inhibitors of PI3K (e.g., apitolisib, copanlisib), AKT (e.g., ipatasertib, perifosine), and mTOR (e.g., sirolimus, temsirolimus) exhibit promising efficacy against various cancers such as Invasive Breast Cancer, Lymphoma, Neoplasms, and Hematological malignancies. This review offers an overview of small molecule inhibitors targeting specific proteins within the RAS upstream and downstream signaling pathways in cancer.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
35
|
Guo Z, Duan Y, Sun K, Zheng T, Liu J, Xu S, Xu J. Advances in SHP2 tunnel allosteric inhibitors and bifunctional molecules. Eur J Med Chem 2024; 275:116579. [PMID: 38889611 DOI: 10.1016/j.ejmech.2024.116579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
SHP2 is a non-receptor tyrosine phosphatase encoded by PTPN11, which performs the functions of regulating cell proliferation, differentiation, apoptosis, and survival through removing tyrosine phosphorylation and modulating various signaling pathways. The overexpression of SHP2 or its mutations is related to developmental diseases and several cancers. Numerous allosteric inhibitors with striking inhibitory potency against SHP2 allosteric pockets have recently been identified, and several SHP2 tunnel allosteric inhibitors have been applied in clinical trials to treat cancers. However, based on clinical results, the efficacy of single-agent treatments has been proven to be suboptimal. Most clinical trials involving SHP2 inhibitors have adopted drug combination strategies. This review briefly discusses the research progress on SHP2 allosteric inhibitors and pathway-dependent drug combination strategies for SHP2 in cancer therapy. In addition, we summarize the current bifunctional molecules of SHP2 and elaborate on the design and structural optimization strategies of these bifunctional molecules in detail, offering further direction for the research on novel SHP2 inhibitors.
Collapse
Affiliation(s)
- Zhichao Guo
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Yiping Duan
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Kai Sun
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Tiandong Zheng
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China
| | - Jie Liu
- Department of Organic Chemistry, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China.
| | - Shengtao Xu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China.
| | - Jinyi Xu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
36
|
Huang M, Zhang F, Zhu Y, Zeng H, Li S. MEST promotes immune escape in gastric cancer by downregulating MHCI expression via SHP2. Int J Biochem Cell Biol 2024; 174:106621. [PMID: 39181599 DOI: 10.1016/j.biocel.2024.106621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Immune escape is a major obstacle to T-cell-based immunotherapy for cancers such as gastric cancer (GC). Mesoderm-specific transcript (MEST) is a tumor-promoting factor that regulates multiple oncogenic signaling pathways. However, the role of MEST-mediated immune escape is unclear. METHODS Bioinformatics analysis of MEST expression and enrichment pathways were performed Quantitative reverse transcription PCR (qPCR) or western blot was used to detect the expression of MEST, Src homology region 2-containing protein tyrosine phosphatase 2 (SHP2), Major histocompatibility class I (MHCI)-related genes. Cell function was assessed by Cell Counting Kit (CCK)-8, Transwell, Lactate dehydrogenase (LDH) kit, flow cytometry, enzyme-linked immunosorbent assay (ELISA), and immunohistochemistry (IHC). Xenograft nude mice and immune-reconstructed mice were used to test the effects of different treatments on tumor growth and immune escape in vivo. RESULTS MEST was upregulated in GC and promoted tumor proliferation, migration, and invasion. Rescue experiments revealed that TNO155 treatment or knockdown of SHP2 promoted the killing ability of CD8+ T cells and the expression of granzyme B (GZMB) and interferon-gamma (IFN-γ), and MEST overexpression reversed the effect. In vivo experiments confirmed that MEST promoted tumor growth, knockdown of MEST inhibited immune escape in GC, and that combination treatment with anti-PD-1 improved anti-tumor activity. CONCLUSION In this study, we demonstrated that MEST inhibited IFN-γ secretion from CD8+ T cells by up-regulating SHP2, thereby downregulating MHCI expression in GC cells to promote immune escape and providing a new T cell-based therapeutic potential for GC.
Collapse
Affiliation(s)
- Min Huang
- Department of Oncology, The First People's Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province 434000, China
| | - Fan Zhang
- Department of Oncology, The First People's Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province 434000, China
| | - Yan Zhu
- Department of Oncology, The First People's Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province 434000, China
| | - Hai Zeng
- Department of Oncology, The First People's Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province 434000, China
| | - Shuang Li
- Department of Oncology, The First People's Hospital Affiliated to Yangtze University, Jingzhou City, Hubei Province 434000, China.
| |
Collapse
|
37
|
Ma C, Kang D, Gao P, Zhang W, Wu X, Xu Z, Han H, Zhang L, Cai Y, Wang Y, Wang Y, Long W. Discovery of JAB-3312, a Potent SHP2 Allosteric Inhibitor for Cancer Treatment. J Med Chem 2024; 67:13534-13549. [PMID: 39110625 DOI: 10.1021/acs.jmedchem.4c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
As an oncogenic phosphatase, SHP2 acts as a converging node in the RTK-RAS-MAPK signaling pathway in cancer cells and suppresses antitumor immunity by passing signals downstream of PD-1. Here, we utilized the extra druggable pocket outside the previously identified SHP2 allosteric tunnel site by the (6,5 fused), 6 spirocyclic system. The optimized compound, JAB-3312, exhibited a SHP2 binding Kd of 0.37 nM, SHP2 enzymatic IC50 of 1.9 nM, KYSE-520 antiproliferative IC50 of 7.4 nM and p-ERK inhibitory IC50 of 0.23 nM. For JAB-3312, an oral dose of 1.0 mg/kg QD was sufficient to achieve 95% TGI in KYSE-520 xenograft model of mouse. JAB-3312 was well-tolerated in animal models, and a close correlation was observed between the plasma concentration of JAB-3312 and the p-ERK inhibition in tumors. Currently, JAB-3312 is undergoing clinical trials as a potential anticancer agent.
Collapse
Affiliation(s)
- Cunbo Ma
- Medicinal Chemistry, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Di Kang
- Pharmacology, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Panliang Gao
- Medicinal Chemistry, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Wei Zhang
- Hits Discovery, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Xinping Wu
- Medicinal Chemistry, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Zilong Xu
- Medicinal Chemistry, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Huifeng Han
- Pharmacology, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Lei Zhang
- Pharmacology, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Yang Cai
- Pharmacology, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Yanping Wang
- Pharmacology, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Yinxiang Wang
- Pharmacology, Jacobio Pharmaceuticals, Beijing 101111, China
| | - Wei Long
- Medicinal Chemistry, Jacobio Pharmaceuticals, Beijing 101111, China
| |
Collapse
|
38
|
Zhu C, Zhao H, Yang W, Chen K, Liu X, Yu Y, Li R, Tan R, Yu Z. Design, Synthesis and Antitumor Activity of a Novel Class of SHP2 Allosteric Inhibitors with a Furanyl Amide-Based Scaffold. J Med Chem 2024. [PMID: 39066713 DOI: 10.1021/acs.jmedchem.4c01217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
SHP2 plays a critical role in modulating tumor growth and PD-1-related signaling pathway, thereby serving as an attractive antitumor target. To date, no antitumor drugs targeting SHP2 have been approved, and hence, the search of SHP2 inhibitors with new chemical scaffolds is urgently needed. Herein, we developed a novel SHP2 allosteric inhibitor SDUY038 with a furanyl amide scaffold, demonstrating potent binding affinity (KD = 0.29 μM), enzymatic activity (IC50 = 1.2 μM) and similar binding interactions to SHP099. At the cellular level, SDUY038 exhibited pan-antitumor activity (IC50 = 7-24 μM) by suppressing pERK expression. Furthermore, SDUY038 significantly inhibited tumor growth in both xenograft and organoid models. Additionally, SDUY038 displayed acceptable bioavailability (F = 14%) and half-life time (t1/2 = 3.95 h). Conclusively, this study introduces the furanyl amide scaffold as a novel class of SHP2 allosteric inhibitors, offering promising lead compounds for further development of new antitumor therapies targeting SHP2.
Collapse
Affiliation(s)
- Chengchun Zhu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P.R. China
| | - Haiyang Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, No. 59, Middle Section of Qinglong Avenue, Mianyang 621010, P.R. China
- Center for Organoids and Translational Pharmacology, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, P.R. China
| | - Wenting Yang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P.R. China
| | - Kai Chen
- Center for New Drug Evaluation, Shandong Academy of Pharmaceutical Sciences, Jinan 250000, P.R. China
| | - Xiaoyu Liu
- Center for New Drug Evaluation, Shandong Academy of Pharmaceutical Sciences, Jinan 250000, P.R. China
| | - Yan Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P.R. China
| | - Rui Li
- Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu 610041, P.R. China
| | - Ruirong Tan
- Center for Organoids and Translational Pharmacology, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, P.R. China
| | - Zhiyi Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P.R. China
| |
Collapse
|
39
|
Liu J, Cao S, Imbach KJ, Gritsenko MA, Lih TSM, Kyle JE, Yaron-Barir TM, Binder ZA, Li Y, Strunilin I, Wang YT, Tsai CF, Ma W, Chen L, Clark NM, Shinkle A, Naser Al Deen N, Caravan W, Houston A, Simin FA, Wyczalkowski MA, Wang LB, Storrs E, Chen S, Illindala R, Li YD, Jayasinghe RG, Rykunov D, Cottingham SL, Chu RK, Weitz KK, Moore RJ, Sagendorf T, Petyuk VA, Nestor M, Bramer LM, Stratton KG, Schepmoes AA, Couvillion SP, Eder J, Kim YM, Gao Y, Fillmore TL, Zhao R, Monroe ME, Southard-Smith AN, Li YE, Jui-Hsien Lu R, Johnson JL, Wiznerowicz M, Hostetter G, Newton CJ, Ketchum KA, Thangudu RR, Barnholtz-Sloan JS, Wang P, Fenyö D, An E, Thiagarajan M, Robles AI, Mani DR, Smith RD, Porta-Pardo E, Cantley LC, Iavarone A, Chen F, Mesri M, Nasrallah MP, Zhang H, Resnick AC, Chheda MG, Rodland KD, Liu T, Ding L. Multi-scale signaling and tumor evolution in high-grade gliomas. Cancer Cell 2024; 42:1217-1238.e19. [PMID: 38981438 PMCID: PMC11337243 DOI: 10.1016/j.ccell.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/12/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024]
Abstract
Although genomic anomalies in glioblastoma (GBM) have been well studied for over a decade, its 5-year survival rate remains lower than 5%. We seek to expand the molecular landscape of high-grade glioma, composed of IDH-wildtype GBM and IDH-mutant grade 4 astrocytoma, by integrating proteomic, metabolomic, lipidomic, and post-translational modifications (PTMs) with genomic and transcriptomic measurements to uncover multi-scale regulatory interactions governing tumor development and evolution. Applying 14 proteogenomic and metabolomic platforms to 228 tumors (212 GBM and 16 grade 4 IDH-mutant astrocytoma), including 28 at recurrence, plus 18 normal brain samples and 14 brain metastases as comparators, reveals heterogeneous upstream alterations converging on common downstream events at the proteomic and metabolomic levels and changes in protein-protein interactions and glycosylation site occupancy at recurrence. Recurrent genetic alterations and phosphorylation events on PTPN11 map to important regulatory domains in three dimensions, suggesting a central role for PTPN11 signaling across high-grade gliomas.
Collapse
Affiliation(s)
- Jingxian Liu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Kathleen J Imbach
- Josep Carreras Leukaemia Research Institute, Badalona, Spain; Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Tung-Shing M Lih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jennifer E Kyle
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Tomer M Yaron-Barir
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Ilya Strunilin
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Yi-Ting Wang
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Chia-Feng Tsai
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lijun Chen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Natalie M Clark
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Andrew Shinkle
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Nataly Naser Al Deen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Wagma Caravan
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Andrew Houston
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Faria Anjum Simin
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Liang-Bo Wang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Erik Storrs
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Siqi Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Ritvik Illindala
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Neurology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Yuping D Li
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Neurology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Reyka G Jayasinghe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Dmitry Rykunov
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sandra L Cottingham
- Department of Pathology, Spectrum Health and Helen DeVos Children's Hospital, Grand Rapids, MI, USA
| | - Rosalie K Chu
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Karl K Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Tyler Sagendorf
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Michael Nestor
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Lisa M Bramer
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Kelly G Stratton
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Athena A Schepmoes
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Sneha P Couvillion
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Josie Eder
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Young-Mo Kim
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Yuqian Gao
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Thomas L Fillmore
- Department of Pathology, Spectrum Health and Helen DeVos Children's Hospital, Grand Rapids, MI, USA
| | - Rui Zhao
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Matthew E Monroe
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Austin N Southard-Smith
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Yang E Li
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rita Jui-Hsien Lu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Jared L Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Maciej Wiznerowicz
- International Institute for Molecular Oncology, Poznań, Poland; Poznan University of Medical Sciences, Poznań, Poland
| | | | | | | | | | - Jill S Barnholtz-Sloan
- Center for Biomedical Informatics and Information Technology & Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20850, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Eunkyung An
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | | | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - D R Mani
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | | | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Antonio Iavarone
- Department of Neurological Surgery and Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Feng Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD 20850, USA
| | - MacLean P Nasrallah
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Adam C Resnick
- Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Milan G Chheda
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Neurology, Washington University in St. Louis, St. Louis, MO 63130, USA.
| | - Karin D Rodland
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR 97221, USA.
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA.
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
40
|
Chouhan S, Sridaran D, Weimholt C, Luo J, Li T, Hodgson MC, Santos LN, Le Sommer S, Fang B, Koomen JM, Seeliger M, Qu CK, Yart A, Kontaridis MI, Mahajan K, Mahajan NP. SHP2 as a primordial epigenetic enzyme expunges histone H3 pTyr-54 to amend androgen receptor homeostasis. Nat Commun 2024; 15:5629. [PMID: 38965223 PMCID: PMC11224269 DOI: 10.1038/s41467-024-49978-4] [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: 04/05/2023] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
Mutations that decrease or increase the activity of the tyrosine phosphatase, SHP2 (encoded by PTPN11), promotes developmental disorders and several malignancies by varying phosphatase activity. We uncovered that SHP2 is a distinct class of an epigenetic enzyme; upon phosphorylation by the kinase ACK1/TNK2, pSHP2 was escorted by androgen receptor (AR) to chromatin, erasing hitherto unidentified pY54-H3 (phosphorylation of histones H3 at Tyr54) epigenetic marks to trigger a transcriptional program of AR. Noonan Syndrome with Multiple Lentigines (NSML) patients, SHP2 knock-in mice, and ACK1 knockout mice presented dramatic increase in pY54-H3, leading to loss of AR transcriptome. In contrast, prostate tumors with high pSHP2 and pACK1 activity exhibited progressive downregulation of pY54-H3 levels and higher AR expression that correlated with disease severity. Overall, pSHP2/pY54-H3 signaling acts as a sentinel of AR homeostasis, explaining not only growth retardation, genital abnormalities and infertility among NSML patients, but also significant AR upregulation in prostate cancer patients.
Collapse
Affiliation(s)
- Surbhi Chouhan
- Department of Surgery, Washington University in St Louis, St Louis, MO, 63110, USA
- 6601, Cancer Research Building, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Dhivya Sridaran
- Department of Surgery, Washington University in St Louis, St Louis, MO, 63110, USA
- 6601, Cancer Research Building, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Cody Weimholt
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Jingqin Luo
- Division of Public Health Sciences, Washington University in St Louis, St Louis, MO, 63110, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Tiandao Li
- Bioinformatics Research Core, Center of Regenerative Medicine, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Myles C Hodgson
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY, 13501, USA
| | - Luana N Santos
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY, 13501, USA
| | - Samantha Le Sommer
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY, 13501, USA
| | - Bin Fang
- Moffitt Cancer Center, SRB3, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - John M Koomen
- Moffitt Cancer Center, SRB3, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Markus Seeliger
- Department of Pharmacological Sciences, Stony Brook University Medical School, BST 7-120, Stony Brook, NY, 11794-8651, USA
| | - Cheng-Kui Qu
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Armelle Yart
- UMR 1301-Inserm 5070-CNRS EFS Univ. P. Sabatier, 4bis Ave Hubert Curien, 31100, Toulouse, France
| | - Maria I Kontaridis
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY, 13501, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kiran Mahajan
- Department of Surgery, Washington University in St Louis, St Louis, MO, 63110, USA
- 6601, Cancer Research Building, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Nupam P Mahajan
- Department of Surgery, Washington University in St Louis, St Louis, MO, 63110, USA.
- 6601, Cancer Research Building, Washington University in St Louis, St Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, 63110, USA.
| |
Collapse
|
41
|
Ortega OO, Ozen M, Wilson BA, Pino JC, Irvin MW, Ildefonso GV, Garbett SP, Lopez CF. Signal execution modes emerge in biochemical reaction networks calibrated to experimental data. iScience 2024; 27:109989. [PMID: 38846004 PMCID: PMC11154230 DOI: 10.1016/j.isci.2024.109989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/29/2024] [Accepted: 05/13/2024] [Indexed: 06/09/2024] Open
Abstract
Mathematical models of biomolecular networks are commonly used to study cellular processes; however, their usefulness to explain and predict dynamic behaviors is often questioned due to the unclear relationship between parameter uncertainty and network dynamics. In this work, we introduce PyDyNo (Python dynamic analysis of biochemical networks), a non-equilibrium reaction-flux based analysis to identify dominant reaction paths within a biochemical reaction network calibrated to experimental data. We first show, in a simplified apoptosis execution model, that despite the thousands of parameter vectors with equally good fits to experimental data, our framework identifies the dynamic differences between these parameter sets and outputs three dominant execution modes, which exhibit varying sensitivity to perturbations. We then apply our methodology to JAK2/STAT5 network in colony-forming unit-erythroid (CFU-E) cells and provide previously unrecognized mechanistic explanation for the survival responses of CFU-E cell population that would have been impossible to deduce with traditional protein-concentration based analyses.
Collapse
Affiliation(s)
- Oscar O. Ortega
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37212, USA
| | - Mustafa Ozen
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37212, USA
- Multiscale Modeling Group, Comp. Bio. Hub, Altos Laboratories, Redwood City, CA 94065, USA
| | - Blake A. Wilson
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37212, USA
| | - James C. Pino
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37212, USA
| | - Michael W. Irvin
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37212, USA
| | - Geena V. Ildefonso
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37212, USA
| | - Shawn P. Garbett
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Carlos F. Lopez
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37212, USA
- Multiscale Modeling Group, Comp. Bio. Hub, Altos Laboratories, Redwood City, CA 94065, USA
| |
Collapse
|
42
|
Pagano L, Pennacchietti V, Malagrinò F, Di Felice M, Toso J, Puglisi E, Gianni S, Toto A. Folding and Binding Kinetics of the Tandem of SH2 Domains from SHP2. Int J Mol Sci 2024; 25:6566. [PMID: 38928272 PMCID: PMC11203950 DOI: 10.3390/ijms25126566] [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: 05/17/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
The SH2 domains of SHP2 play a crucial role in determining the function of the SHP2 protein. While the folding and binding properties of the isolated NSH2 and CSH2 domains have been extensively studied, there is limited information about the tandem SH2 domains. This study aims to elucidate the folding and binding kinetics of the NSH2-CSH2 tandem domains of SHP2 through rapid kinetic experiments, complementing existing data on the isolated domains. The results indicate that while the domains generally fold and unfold independently, acidic pH conditions induce complex scenarios involving the formation of a misfolded intermediate. Furthermore, a comparison of the binding kinetics of isolated NSH2 and CSH2 domains with the NSH2-CSH2 tandem domains, using peptides that mimic specific portions of Gab2, suggests a dynamic interplay between NSH2 and CSH2 in binding Gab2 that modulate the microscopic association rate constant of the binding reaction. These findings, discussed in the context of previous research on the NSH2 and CSH2 domains, enhance our understanding of the function of the SH2 domain tandem of SHP2.
Collapse
Affiliation(s)
- Livia Pagano
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma, 00185 Rome, Italy; (L.P.); (V.P.); (M.D.F.); (J.T.); (E.P.); (S.G.)
| | - Valeria Pennacchietti
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma, 00185 Rome, Italy; (L.P.); (V.P.); (M.D.F.); (J.T.); (E.P.); (S.G.)
| | - Francesca Malagrinò
- Dipartimento di Medicina Clinica, Sanità Pubblica, Scienze della Vita e Dell’ambiente, Università dell’Aquila, Piazzale Salvatore Tommasi 1, Coppito, 67010 L’Aquila, Italy;
| | - Mariana Di Felice
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma, 00185 Rome, Italy; (L.P.); (V.P.); (M.D.F.); (J.T.); (E.P.); (S.G.)
| | - Julian Toso
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma, 00185 Rome, Italy; (L.P.); (V.P.); (M.D.F.); (J.T.); (E.P.); (S.G.)
| | - Elena Puglisi
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma, 00185 Rome, Italy; (L.P.); (V.P.); (M.D.F.); (J.T.); (E.P.); (S.G.)
| | - Stefano Gianni
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma, 00185 Rome, Italy; (L.P.); (V.P.); (M.D.F.); (J.T.); (E.P.); (S.G.)
| | - Angelo Toto
- Dipartimento di Scienze Biochimiche “A. Rossi Fanelli”, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza Università di Roma, 00185 Rome, Italy; (L.P.); (V.P.); (M.D.F.); (J.T.); (E.P.); (S.G.)
| |
Collapse
|
43
|
Miao Y, Bai Y, Miao J, Murray AA, Lin J, Dong J, Qu Z, Zhang RY, Nguyen QD, Wang S, Yu J, Nguele Meke F, Zhang ZY. Off-target autophagy inhibition by SHP2 allosteric inhibitors contributes to their antitumor activity in RAS-driven cancers. J Clin Invest 2024; 134:e177142. [PMID: 38842946 PMCID: PMC11291269 DOI: 10.1172/jci177142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 06/04/2024] [Indexed: 08/02/2024] Open
Abstract
Aberrant activation of RAS/MAPK signaling is common in cancer, and efforts to inhibit pathway components have yielded drugs with promising clinical activities. Unfortunately, treatment-provoked adaptive resistance mechanisms inevitably develop, limiting their therapeutic potential. As a central node essential for receptor tyrosine kinase-mediated RAS activation, SHP2 has emerged as an attractive cancer target. Consequently, many SHP2 allosteric inhibitors are now in clinical testing. Here we discovered a previously unrecognized off-target effect associated with SHP2 allosteric inhibitors. We found that these inhibitors accumulate in the lysosome and block autophagic flux in an SHP2-independent manner. We showed that off-target autophagy inhibition by SHP2 allosteric inhibitors contributes to their antitumor activity. We also demonstrated that SHP2 allosteric inhibitors harboring this off-target activity not only suppress oncogenic RAS signaling but also overcome drug resistance such as MAPK rebound and protective autophagy in response to RAS/MAPK pathway blockage. Finally, we exemplified a therapeutic framework that harnesses both the on- and off-target activities of SHP2 allosteric inhibitors for improved treatment of mutant RAS-driven and drug-resistant malignancies such as pancreatic and colorectal cancers.
Collapse
Affiliation(s)
- Yiming Miao
- Department of Medicinal Chemistry and Molecular Pharmacology and
| | - Yunpeng Bai
- Department of Medicinal Chemistry and Molecular Pharmacology and
| | - Jinmin Miao
- Department of Medicinal Chemistry and Molecular Pharmacology and
| | | | - Jianping Lin
- Department of Medicinal Chemistry and Molecular Pharmacology and
| | - Jiajun Dong
- Department of Medicinal Chemistry and Molecular Pharmacology and
| | - Zihan Qu
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| | - Ruo-Yu Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology and
| | - Quyen D. Nguyen
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| | - Shaomeng Wang
- Departments of Internal Medicine, Pharmacology, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jingmei Yu
- Department of Medicinal Chemistry and Molecular Pharmacology and
| | | | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology and
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
- Institute for Cancer Research and
- Institute for Drug Discovery, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
44
|
Rahmat JN, Liu J, Chen T, Li Z, Zhang Y. Engineered biological nanoparticles as nanotherapeutics for tumor immunomodulation. Chem Soc Rev 2024; 53:5862-5903. [PMID: 38716589 DOI: 10.1039/d3cs00602f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Biological nanoparticles, or bionanoparticles, are small molecules manufactured in living systems with complex production and assembly machinery. The products of the assembly systems can be further engineered to generate functionalities for specific purposes. These bionanoparticles have demonstrated advantages such as immune system evasion, minimal toxicity, biocompatibility, and biological clearance. Hence, bionanoparticles are considered the new paradigm in nanoscience research for fabricating safe and effective nanoformulations for therapeutic purposes. Harnessing the power of the immune system to recognize and eradicate malignancies is a viable strategy to achieve better therapeutic outcomes with long-term protection from disease recurrence. However, cancerous tissues have evolved to become invisible to immune recognition and to transform the tumor microenvironment into an immunosuppressive dwelling, thwarting the immune defense systems and creating a hospitable atmosphere for cancer growth and progression. Thus, it is pertinent that efforts in fabricating nanoformulations for immunomodulation are mindful of the tumor-induced immune aberrations that could render cancer nanotherapy inoperable. This review systematically categorizes the immunosuppression mechanisms, the regulatory immunosuppressive cellular players, and critical suppressive molecules currently targeted as breakthrough therapies in the clinic. Finally, this review will summarize the engineering strategies for affording immune moderating functions to bionanoparticles that tip the tumor microenvironment (TME) balance toward cancer elimination, a field still in the nascent stage.
Collapse
Affiliation(s)
- Juwita N Rahmat
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117585, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Taili Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - ZhiHong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yong Zhang
- Department of Biomedical Engineering, College of Engineering, The City University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
45
|
Pandey G, Mazzacurati L, Rowsell TM, Horvat NP, Amin NE, Zhang G, Akuffo AA, Colin-Leitzinger CM, Haura EB, Kuykendall AT, Zhang L, Epling-Burnette PK, Reuther GW. SHP2 inhibition displays efficacy as a monotherapy and in combination with JAK2 inhibition in preclinical models of myeloproliferative neoplasms. Am J Hematol 2024; 99:1040-1055. [PMID: 38440831 PMCID: PMC11096011 DOI: 10.1002/ajh.27282] [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: 09/26/2023] [Revised: 01/30/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024]
Abstract
Myeloproliferative neoplasms (MPNs), including polycythemia vera, essential thrombocytosis, and primary myelofibrosis, are clonal hematopoietic neoplasms driven by mutationally activated signaling by the JAK2 tyrosine kinase. Although JAK2 inhibitors can improve MPN patients' quality of life, they do not induce complete remission as disease-driving cells persistently survive therapy. ERK activation has been highlighted as contributing to JAK2 inhibitor persistent cell survival. As ERK is a component of signaling by activated RAS proteins and by JAK2 activation, we sought to inhibit RAS activation to enhance responses to JAK2 inhibition in preclinical MPN models. We found the SHP2 inhibitor RMC-4550 significantly enhanced growth inhibition of MPN cell lines in combination with the JAK2 inhibitor ruxolitinib, effectively preventing ruxolitinib persistent growth, and the growth and viability of established ruxolitinib persistent cells remained sensitive to SHP2 inhibition. Both SHP2 and JAK2 inhibition diminished cellular RAS-GTP levels, and their concomitant inhibition enhanced ERK inactivation and increased apoptosis. Inhibition of SHP2 inhibited the neoplastic growth of MPN patient hematopoietic progenitor cells and exhibited synergy with ruxolitinib. RMC-4550 antagonized MPN phenotypes and increased survival of an MPN mouse model driven by MPL-W515L. The combination of RMC-4550 and ruxolitinib, which was safe and tolerated in healthy mice, further inhibited disease compared to ruxolitinib monotherapy, including extending survival. Given SHP2 inhibitors are undergoing clinical evaluation in patients with solid tumors, our preclinical findings suggest that SHP2 is a candidate therapeutic target with potential for rapid translation to clinical assessment to improve current targeted therapies for MPN patients.
Collapse
Affiliation(s)
- Garima Pandey
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
| | - Lucia Mazzacurati
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
| | - Tegan M. Rowsell
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
| | | | - Narmin E. Amin
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
| | - Guolin Zhang
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL USA
| | - Afua A. Akuffo
- Department of Immunology, Moffitt Cancer Center, Tampa, FL USA
| | | | - Eric B. Haura
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL USA
| | | | - Ling Zhang
- Department of Pathology, Moffitt Cancer Center, Tampa, FL USA
| | | | - Gary W. Reuther
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL USA
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL USA
| |
Collapse
|
46
|
Ma D, Yang M, Sun C, Cui X, Xiong G, Wang Q, Jing W, Chen H, Lv X, Liu S, Li T, Zhao Y, Han L. cGAS suppresses hepatocellular carcinoma independent of its cGAMP synthase activity. Cell Death Differ 2024; 31:722-737. [PMID: 38594443 PMCID: PMC11164996 DOI: 10.1038/s41418-024-01291-9] [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: 05/26/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) is a key innate immune sensor that recognizes cytosolic DNA to induce immune responses against invading pathogens. The role of cGAS is conventionally recognized as a nucleotidyltransferase to catalyze the synthesis of cGAMP upon recognition of cytosolic DNA, which leads to the activation of STING and production of type I/III interferon to fight against the pathogen. However, given that hepatocytes are lack of functional STING expression, it is intriguing to define the role of cGAS in hepatocellular carcinoma (HCC), the liver parenchymal cells derived malignancy. In this study, we revealed that cGAS was significantly downregulated in clinical HCC tissues, and its dysregulation contributed to the progression of HCC. We further identified cGAS as an immune tyrosine inhibitory motif (ITIM) containing protein, and demonstrated that cGAS inhibited the progression of HCC and increased the response of HCC to sorafenib treatment by suppressing PI3K/AKT/mTORC1 pathway in cellular and animal models. Mechanistically, cGAS recruits SH2-containing tyrosine phosphatase 1 (SHP1) via ITIM, and dephosphorylates p85 in phosphatidylinositol 3-kinase (PI3K), which leads to the suppression of AKT-mTORC1 pathway. Thus, cGAS is identified as a novel tumor suppressor in HCC via its function independent of its conventional role as cGAMP synthase, which indicates a novel therapeutic strategy for advanced HCC by modulating cGAS signaling.
Collapse
Affiliation(s)
- Dapeng Ma
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Min Yang
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Caiyu Sun
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiuling Cui
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gaozhong Xiong
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiushi Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Haiqiang Chen
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoting Lv
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shili Liu
- Department of Microbiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Li
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yunxue Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lihui Han
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
47
|
Scheiter A, Lu LC, Gao LH, Feng GS. Complex Roles of PTPN11/SHP2 in Carcinogenesis and Prospect of Targeting SHP2 in Cancer Therapy. ANNUAL REVIEW OF CANCER BIOLOGY 2024; 8:15-33. [PMID: 39959686 PMCID: PMC11824402 DOI: 10.1146/annurev-cancerbio-062722-013740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
The non-receptor tyrosine phosphatase SHP2 has been at the center of cell signaling research for three decades. SHP2 is required to fully activate the RTK-RAS-ERK cascade, although the underlying mechanisms are not completely understood. PTPN11, coding for SHP2, is the first identified proto-oncogene that encodes a tyrosine phosphatase, with dominantly activating mutations detected in leukemias and solid tumors. However, SHP2 has been shown to have pro- and anti-oncogenic effects, and the most recent data reveal opposite activities of SHP2 in tumor cells and microenvironment cells. Allosteric SHP2 inhibitors show promising anti-tumor effects and overcome resistance to inhibitors of RAS-ERK signaling in animal models. Many clinical trials with orally bioactive SHP2 inhibitors, alone or combined with other regimens, are ongoing for a variety of cancers worldwide, with therapeutic outcomes yet unknown. This review discusses the multi-faceted SHP2 functions in oncogenesis, preclinical studies and clinical trials with SHP2 inhibitors in oncological treatment.
Collapse
Affiliation(s)
- Alexander Scheiter
- Department of Pathology, and Moores Cancer Center, School of Medicine, University of California San Diego, La Jolla, California 92093
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Li-Chun Lu
- Department of Pathology, and Moores Cancer Center, School of Medicine, University of California San Diego, La Jolla, California 92093
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan (ROC)
| | - Lilian H. Gao
- Department of Pathology, and Moores Cancer Center, School of Medicine, University of California San Diego, La Jolla, California 92093
- Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, California 92093
| | - Gen-Sheng Feng
- Department of Pathology, and Moores Cancer Center, School of Medicine, University of California San Diego, La Jolla, California 92093
- Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, California 92093
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
48
|
Yang D, Jian Z, Tang C, Chen Z, Zhou Z, Zheng L, Peng X. Zebrafish Congenital Heart Disease Models: Opportunities and Challenges. Int J Mol Sci 2024; 25:5943. [PMID: 38892128 PMCID: PMC11172925 DOI: 10.3390/ijms25115943] [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: 04/14/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Congenital heart defects (CHDs) are common human birth defects. Genetic mutations potentially cause the exhibition of various pathological phenotypes associated with CHDs, occurring alone or as part of certain syndromes. Zebrafish, a model organism with a strong molecular conservation similar to humans, is commonly used in studies on cardiovascular diseases owing to its advantageous features, such as a similarity to human electrophysiology, transparent embryos and larvae for observation, and suitability for forward and reverse genetics technology, to create various economical and easily controlled zebrafish CHD models. In this review, we outline the pros and cons of zebrafish CHD models created by genetic mutations associated with single defects and syndromes and the underlying pathogenic mechanism of CHDs discovered in these models. The challenges of zebrafish CHD models generated through gene editing are also discussed, since the cardiac phenotypes resulting from a single-candidate pathological gene mutation in zebrafish might not mirror the corresponding human phenotypes. The comprehensive review of these zebrafish CHD models will facilitate the understanding of the pathogenic mechanisms of CHDs and offer new opportunities for their treatments and intervention strategies.
Collapse
|
49
|
Lee S, Kim J, Ryu HH, Jang H, Lee D, Lee S, Song JM, Lee YS, Ho Suh Y. SHP2 regulates GluA2 tyrosine phosphorylation required for AMPA receptor endocytosis and mGluR-LTD. Proc Natl Acad Sci U S A 2024; 121:e2316819121. [PMID: 38657042 PMCID: PMC11066993 DOI: 10.1073/pnas.2316819121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024] Open
Abstract
Posttranslational modifications regulate the properties and abundance of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors that mediate fast excitatory synaptic transmission and synaptic plasticity in the central nervous system. During long-term depression (LTD), protein tyrosine phosphatases (PTPs) dephosphorylate tyrosine residues in the C-terminal tail of AMPA receptor GluA2 subunit, which is essential for GluA2 endocytosis and group I metabotropic glutamate receptor (mGluR)-dependent LTD. However, as a selective downstream effector of mGluRs, the mGluR-dependent PTP responsible for GluA2 tyrosine dephosphorylation remains elusive at Schaffer collateral (SC)-CA1 synapses. In the present study, we find that mGluR5 stimulation activates Src homology 2 (SH2) domain-containing phosphatase 2 (SHP2) by increasing phospho-Y542 levels in SHP2. Under steady-state conditions, SHP2 plays a protective role in stabilizing phospho-Y869 of GluA2 by directly interacting with GluA2 phosphorylated at Y869, without affecting GluA2 phospho-Y876 levels. Upon mGluR5 stimulation, SHP2 dephosphorylates GluA2 at Y869 and Y876, resulting in GluA2 endocytosis and mGluR-LTD. Our results establish SHP2 as a downstream effector of mGluR5 and indicate a dual action of SHP2 in regulating GluA2 tyrosine phosphorylation and function. Given the implications of mGluR5 and SHP2 in synaptic pathophysiology, we propose SHP2 as a promising therapeutic target for neurodevelopmental and autism spectrum disorders.
Collapse
Affiliation(s)
- Sanghyeon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Jungho Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Hyun-Hee Ryu
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Hanbyul Jang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - DoEun Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Seungha Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Jae-man Song
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Yong-Seok Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Young Ho Suh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| |
Collapse
|
50
|
Gao Y, Xing S, Hu L. Probing the Immunoreceptor Tyrosine-Based Inhibition Motif Interaction Protein Partners with Proteomics. Molecules 2024; 29:1977. [PMID: 38731468 PMCID: PMC11085718 DOI: 10.3390/molecules29091977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Phosphorylation of tyrosine is the basic mode of protein function and signal transduction in organisms. This process is regulated by protein tyrosine kinases (PTKs) and protein tyrosinases (PTPs). Immunoreceptor tyrosine-based inhibition motif (ITIM) has been considered as regulating the PTP activity through the interaction with the partner proteins in the cell signal pathway. The ITIM sequences need to be phosphorylated first to active the downstream signaling proteins. To explore potential regulatory mechanisms, the ITIM sequences of two transmembrane immunoglobulin proteins, myelin P0 protein-related protein (PZR) and programmed death 1 (PD-1), were analyzed to investigate their interaction with proteins involved in regulatory pathways. We discovered that phosphorylated ITIM sequences can selectively interact with the tyrosine phosphatase SHP2. Specifically, PZR-N-ITIM (pY) may be critical in the interaction between the ITIM and SH2 domains of SHP2, while PD1-C-ITSM (pY) may play a key role in the interaction between the ITIM and SH2 domains of SHP2. Quite a few proteins were identified containing the SH2 domain, exhibiting phosphorylation-mediated interaction with PZR-ITIM. In this study, 14 proteins with SH2 structural domains were identified by GO analysis on 339 proteins associated to the affinity pull-down of PZR-N-ITIM (pY). Through the SH2 domains, these proteins may interact with PZR-ITIM in a phosphorylation-dependent manner.
Collapse
Affiliation(s)
| | - Shu Xing
- School of Life Sciences, Jilin University, Changchun 130012, China;
| | - Lianghai Hu
- School of Life Sciences, Jilin University, Changchun 130012, China;
| |
Collapse
|