1
|
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
|
2
|
Song Y, Ren X, Yang X, Xiong J, Wang W, Tang K, Yu B. Structure-Guided Design of Pyrazolopyrimidinones as Highly Potent and Selective Allosteric SHP2 Inhibitors. J Med Chem 2025. [PMID: 40235316 DOI: 10.1021/acs.jmedchem.5c00524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Src homology-2-containing protein tyrosine phosphatase 2 (SHP2) plays crucial roles in various biological processes and has become a promising target for cancer therapy. In this work, we presented the structure-guided design of new allosteric SHP2 inhibitors, leading to the identification of the pyrazolopyrimidinone derivatives TK-684 and TK-685. Both compounds were highly potent and selective allosteric SHP2 inhibitors (TK-684: SHP2WT IC50 = 2.1 nM; Ki = 0.89 nM; TK-685: SHP2WT IC50 = 1.5 nM; Ki = 0.87 nM), likely binding to the "tunnel" allosteric site of SHP2. By targeting SHP2-mediated AKT and ERK signaling pathways, TK-684 and TK-685 suppressed cell proliferation and induced apoptosis in esophageal cancer cells. Additionally, oral administration of TK-685 demonstrated good antitumor effects in the KYSE-150 xenograft mouse model, with a T/C value of 76.8%. Collectively, the pyrazolopyrimidinone derivatives represent promising lead compounds for the treatment of esophageal cancer, where SHP2 is dysregulated.
Collapse
Affiliation(s)
- Yihui Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou 350122, China
| | - Xiangli Ren
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xinyu Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jinbo Xiong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenwen Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kai Tang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Yu
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer, College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
3
|
Pires LVL, Da Cás E, de Melo LC, Nakaie CMA, Aiello VD, Yamamoto GL, Honjo RS, Kim CA, Bertola DR. Plastic Bronchitis in Noonan Syndrome: Further Evidence Suggesting a Higher Risk of Lymphatic Abnormalities in Individuals Harboring Variants in PTPN11 Residue p.Phe285. Mol Syndromol 2025; 16:165-170. [PMID: 40176838 PMCID: PMC11961087 DOI: 10.1159/000540570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/24/2024] [Indexed: 01/17/2025] Open
Abstract
INTRODUCTION Noonan syndrome (NS) is a Mendelian phenotype, member of the RASopathies, a group of clinically overlapping multisystem disorders caused by germline variants in the RAS-MAPK signaling pathway genes. Among the clinical findings in NS, lymphatic abnormalities (LAs) are diagnosed in approximately 30%, mostly in individuals harboring variants in RIT1 and SOS2. This genotype-phenotype correlation is not precise, and recent evidence suggests a higher prevalence of LAs in individuals harboring variants on p.Phe285 residue in PTPN11, the main gene responsible for NS. CASE PRESENTATION Here, we report a novel case of NS harboring the PTPN11:p.Phe285Ser variant that evolved with chylothorax and presented the rare finding of plastic bronchitis, an uncommon and underdiagnosed pulmonary disease, characterized by production of cohesive and branching casts filling the airways. We also provide a review of other individuals with NS and LA harboring variants on Phe285 residue in PTPN11 from our service and from the literature and compared its prevalence with the most commonly affected residue in PTPN11-related NS (p.Asn308), which indicated that variants in the p.Phe285 residue might predispose to LA. CONCLUSION We suggest that, when this variant is identified in an individual, clinicians should be warned of a possible higher prevalence of LA and a prompt evaluation should be performed if any clinical signs are noticed.
Collapse
Affiliation(s)
- Lucas Vieira Lacerda Pires
- Unidade de Genética, Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Eduardo Da Cás
- Unidade de Genética, Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Letícia Cole de Melo
- Unidade de Pneumologia, Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Cleyde Mirian Aversa Nakaie
- Unidade de Pneumologia, Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Vera Dermachi Aiello
- Laboratório de Anatomia Patológica, Instituto do Coração, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Guilherme Lopes Yamamoto
- Unidade de Genética, Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- DASA, São Paulo, Brazil
- Centro de Pesquisa sobre o Genoma Humano e Células Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Rachel Sayuri Honjo
- Unidade de Genética, Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Chong Ae Kim
- Unidade de Genética, Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Débora Romeo Bertola
- Unidade de Genética, Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Centro de Pesquisa sobre o Genoma Humano e Células Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
4
|
Kim SH, Bulos ML, Adams JA, Yun BK, Bishop AC. Single Ion Pair Is Essential for Stabilizing SHP2's Open Conformation. Biochemistry 2024; 63:273-281. [PMID: 38251939 DOI: 10.1021/acs.biochem.3c00609] [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: 01/23/2024]
Abstract
Src-homology-2-domain-containing PTP-2 (SHP2) is a widely expressed signaling enzyme whose misregulation is associated with multiple human pathologies. SHP2's enzymatic activity is controlled by a conformational equilibrium between its autoinhibited ("closed") state and its activated ("open") state. Although SHP2's closed state has been extensively characterized, the putative structure of its open form has only been revealed in the context of a highly activated mutant (E76K), and no systematic studies of the biochemical determinants of SHP2's open-state stabilization have been reported. To identify amino-acid interactions that are critical for stabilizing SHP2's active state, we carried out a mutagenic study of residues that lie at potentially important interdomain interfaces of the open conformation. The open/closed equilibria of the mutants were evaluated, and we identified several interactions that contribute to the stabilization of SHP2's open state. In particular, our findings establish that an ion pair between glutamate 249 on SHP2's PTP domain and arginine 111 on an interdomain loop is the key determinant of SHP2's open-state stabilization. Mutations that disrupt the R111/E249 ion pair substantially shift SHP2's open/closed equilibrium to the closed state, even compared to wild-type SHP2's basal-state equilibrium, which strongly favors the closed state. To the best of our knowledge, the ion-pair variants uncovered in this study are the first known SHP2 mutants in which autoinhibition is augmented with respect to the wild-type protein. Such "hyperinhibited" mutants may provide useful tools for signaling studies that investigate the connections between SHP2 inhibition and the suppression of human disease progression.
Collapse
Affiliation(s)
- Sean H Kim
- Department of Chemistry and Program in Biochemistry & Biophysics, Amherst College, Amherst, Massachusetts 01002, United States
| | - Maya L Bulos
- Department of Chemistry and Program in Biochemistry & Biophysics, Amherst College, Amherst, Massachusetts 01002, United States
| | - Jennifer A Adams
- Department of Chemistry and Program in Biochemistry & Biophysics, Amherst College, Amherst, Massachusetts 01002, United States
| | - B Koun Yun
- Department of Chemistry and Program in Biochemistry & Biophysics, Amherst College, Amherst, Massachusetts 01002, United States
| | - Anthony C Bishop
- Department of Chemistry and Program in Biochemistry & Biophysics, Amherst College, Amherst, Massachusetts 01002, United States
| |
Collapse
|
5
|
Guo M, Li Z, Gu M, Gu J, You Q, Wang L. Targeting phosphatases: From molecule design to clinical trials. Eur J Med Chem 2024; 264:116031. [PMID: 38101039 DOI: 10.1016/j.ejmech.2023.116031] [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: 10/23/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
Phosphatase is a kind of enzyme that can dephosphorylate target proteins, which can be divided into serine/threonine phosphatase and tyrosine phosphatase according to its mode of action. Current evidence showed multiple phosphatases were highly correlated with diseases including various cancers, demonstrating them as potential targets. However, currently, targeting phosphatases with small molecules faces many challenges, resulting in no drug approved. In this case, phosphatases are even regarded as "undruggable" targets for a long time. Recently, a variety of strategies have been adopted in the design of small molecule inhibitors targeting phosphatases, leading many of them to enter into the clinical trials. In this review, we classified these inhibitors into 4 types, including (1) molecular glues, (2) small molecules targeting catalytic sites, (3) allosteric inhibition, and (4) bifunctional molecules (proteolysis targeting chimeras, PROTACs). These molecules with diverse strategies prove the feasibility of phosphatases as drug targets. In addition, the combination therapy of phosphatase inhibitors with other drugs has also entered clinical trials, which suggests a broad prospect. Thus, targeting phosphatases with small molecules by different strategies is emerging as a promising way in the modulation of pathogenetic phosphorylation.
Collapse
Affiliation(s)
- Mochen Guo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zekun Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Mingxiao Gu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Junrui Gu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
6
|
Liu Y, Zhang W, Jang H, Nussinov R. SHP2 clinical phenotype, cancer, or RASopathies, can be predicted by mutant conformational propensities. Cell Mol Life Sci 2023; 81:5. [PMID: 38085330 PMCID: PMC11072105 DOI: 10.1007/s00018-023-05052-8] [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: 07/12/2023] [Revised: 10/20/2023] [Accepted: 11/11/2023] [Indexed: 12/18/2023]
Abstract
SHP2 phosphatase promotes full activation of the RTK-dependent Ras/MAPK pathway. Its mutations can drive cancer and RASopathies, a group of neurodevelopmental disorders (NDDs). Here we ask how same residue mutations in SHP2 can lead to both cancer and NDD phenotypes, and whether we can predict what the outcome will be. We collected and analyzed mutation data from the literature and cancer databases and performed molecular dynamics simulations of SHP2 mutants. We show that both cancer and Noonan syndrome (NS, a RASopathy) mutations favor catalysis-prone conformations. As to cancer versus RASopathies, we demonstrate that cancer mutations are more likely to accelerate SHP2 activation than the NS mutations at the same genomic loci, in line with NMR data for K-Ras4B more aggressive mutations. The compiled experimental data and dynamic features of SHP2 mutants lead us to propose that different from strong oncogenic mutations, SHP2 activation by NS mutations is less likely to induce a transition of the ensemble from the SHP2 inactive state to the active state. Strong signaling promotes cell proliferation, a hallmark of cancer. Weak, or moderate signals are associated with differentiation. In embryonic neural cells, dysregulated differentiation is connected to NDDs. Our innovative work offers structural guidelines for identifying and correlating mutations with clinical outcomes, and an explanation for why bearers of RASopathy mutations may have a higher probability of cancer. Finally, we propose a drug strategy against SHP2 variants-promoting cancer and RASopathies.
Collapse
Affiliation(s)
- Yonglan Liu
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Wengang Zhang
- Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA.
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
| |
Collapse
|
7
|
Segura J, Rose Y, Bi C, Duarte J, Burley SK, Bittrich S. RCSB Protein Data Bank: visualizing groups of experimentally determined PDB structures alongside computed structure models of proteins. FRONTIERS IN BIOINFORMATICS 2023; 3:1311287. [PMID: 38111685 PMCID: PMC10726007 DOI: 10.3389/fbinf.2023.1311287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/17/2023] [Indexed: 12/20/2023] Open
Abstract
Recent advances in Artificial Intelligence and Machine Learning (e.g., AlphaFold, RosettaFold, and ESMFold) enable prediction of three-dimensional (3D) protein structures from amino acid sequences alone at accuracies comparable to lower-resolution experimental methods. These tools have been employed to predict structures across entire proteomes and the results of large-scale metagenomic sequence studies, yielding an exponential increase in available biomolecular 3D structural information. Given the enormous volume of this newly computed biostructure data, there is an urgent need for robust tools to manage, search, cluster, and visualize large collections of structures. Equally important is the capability to efficiently summarize and visualize metadata, biological/biochemical annotations, and structural features, particularly when working with vast numbers of protein structures of both experimental origin from the Protein Data Bank (PDB) and computationally-predicted models. Moreover, researchers require advanced visualization techniques that support interactive exploration of multiple sequences and structural alignments. This paper introduces a suite of tools provided on the RCSB PDB research-focused web portal RCSB. org, tailor-made for efficient management, search, organization, and visualization of this burgeoning corpus of 3D macromolecular structure data.
Collapse
Affiliation(s)
- Joan Segura
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, San Diego Supercomputer Center, University of California San Diego, San Diego, CA, United States
| | - Yana Rose
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, San Diego Supercomputer Center, University of California San Diego, San Diego, CA, United States
| | - Chunxiao Bi
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, San Diego Supercomputer Center, University of California San Diego, San Diego, CA, United States
| | - Jose Duarte
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, San Diego Supercomputer Center, University of California San Diego, San Diego, CA, United States
| | - Stephen K. Burley
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, San Diego Supercomputer Center, University of California San Diego, San Diego, CA, United States
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Sebastian Bittrich
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, San Diego Supercomputer Center, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
8
|
Serbina A, Bishop AC. Quantitation of autoinhibitory defects in pathogenic SHP2 mutants by differential scanning fluorimetry. Anal Biochem 2023; 680:115300. [PMID: 37659706 PMCID: PMC10530186 DOI: 10.1016/j.ab.2023.115300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/26/2023] [Accepted: 08/25/2023] [Indexed: 09/04/2023]
Abstract
Src-homology-2-domain-containing protein tyrosine phosphatase-2 (SHP2) is a signaling enzyme whose activity is governed by an equilibrium between autoinhibited and activated states. Regulation of SHP2 activity is critical for cellular homeostasis, and mutations that alter its autoregulatory equilibrium cause cancers and developmental disorders. Several methods for assessing the strength of autoinhibitory interactions in SHP2 mutants have been previously reported, but each has limitations. We show that differential scanning fluorimetry provides a rapid, quantitative measure of SHP2 autoinhibition that is independent of the intrinsic activity of the SHP2 mutant being analyzed, does not involve protein labeling, and does not require specialized instrumentation.
Collapse
Affiliation(s)
- Anna Serbina
- Amherst College, Department of Chemistry, Amherst, MA, 01002, United States
| | - Anthony C Bishop
- Amherst College, Department of Chemistry, Amherst, MA, 01002, United States.
| |
Collapse
|
9
|
Lade D, Agazie YM. Targeting SHP2 with an Active Site Inhibitor Blocks Signaling and Breast Cancer Cell Phenotypes. ACS BIO & MED CHEM AU 2023; 3:418-428. [PMID: 37876496 PMCID: PMC10591299 DOI: 10.1021/acsbiomedchemau.3c00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 10/26/2023]
Abstract
The Src homology phosphotyrosyl phosphatase 2 (SHP2) is an oncogenic protein for which targeted therapies are being sought. In line with this idea, we have previously reported the development of a specific active site inhibitor named CNBDA that showed effectivity in suppressing the transformation phenotypes of breast cancer cells. To improve efficacy, we introduced limited modifications to the parent compound and tested potency in vitro and under cell culture conditions. Of these modifications, removal of one of the butyric acid groups led to the production of a compound named CNBCA, which showed a 5.7-fold better potency against the SHP2 enzyme activity in vitro. In addition, CNBCA showed better selectivity to SHP2 than the control PTPs (SHP1 and PTP1B) as determined by the phosphatase assay. Furthermore, CNBCA binds and inhibits enzyme activity of full-length SHP2 in cellular contexts, downregulates SHP2 mediated signaling, and suppresses breast cancer cell phenotypes, including cell proliferation, colony formation, and mammosphere growth. These findings show that targeting SHP2 with CNBCA is effective against the cancerous properties of breast cancer cells.
Collapse
Affiliation(s)
- Dhanaji
M. Lade
- One Medical Center Drive, Department
of Biochemistry and Molecular Medicine, School of Medicine, West Virginia University, P.O. Box 9142, Morgantown, West Virginia 26506, United States
| | - Yehenew M. Agazie
- One Medical Center Drive, Department
of Biochemistry and Molecular Medicine, School of Medicine, West Virginia University, P.O. Box 9142, Morgantown, West Virginia 26506, United States
| |
Collapse
|
10
|
Xie X, Yu T, Li X, Zhang N, Foster LJ, Peng C, Huang W, He G. Recent advances in targeting the "undruggable" proteins: from drug discovery to clinical trials. Signal Transduct Target Ther 2023; 8:335. [PMID: 37669923 PMCID: PMC10480221 DOI: 10.1038/s41392-023-01589-z] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/22/2023] [Accepted: 08/02/2023] [Indexed: 09/07/2023] Open
Abstract
Undruggable proteins are a class of proteins that are often characterized by large, complex structures or functions that are difficult to interfere with using conventional drug design strategies. Targeting such undruggable targets has been considered also a great opportunity for treatment of human diseases and has attracted substantial efforts in the field of medicine. Therefore, in this review, we focus on the recent development of drug discovery targeting "undruggable" proteins and their application in clinic. To make this review well organized, we discuss the design strategies targeting the undruggable proteins, including covalent regulation, allosteric inhibition, protein-protein/DNA interaction inhibition, targeted proteins regulation, nucleic acid-based approach, immunotherapy and others.
Collapse
Affiliation(s)
- Xin Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Tingting Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| | - Xiang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
- Department of Dermatology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China.
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China.
| | - Gu He
- Department of Dermatology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
11
|
Design and synthesis of improved active-site SHP2 inhibitors with anti-breast cancer cell effects. Eur J Med Chem 2023; 247:115017. [PMID: 36584630 DOI: 10.1016/j.ejmech.2022.115017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
The Src homology containing phosphotyrosyl phosphatase 2 (SHP2) is a bona fide oncogene particularly in cancers driven by overexpression of receptor tyrosine kinases (RTKs). As such, there is a growing interest to target SHP2 in cancer. Based on these premises, several active site (type I) and allosteric site (type II) inhibitors have been developed, but no SHP2 targeting therapies have reached the clinic yet. In an effort to fill these gaps, we embarked on producing optimized versions of our parent active-site SHP2 inhibitor CNBDA. The objectives were to produce derivatives with increased inhibitory potential and improved selectivity. Accordingly, we designed derivatives around the CNBDA scaffold and predicted their binding property by in silico molecular modeling. Based on comparative differences in free energy of binding to the SHP2 versus the SHP1 active sites, ten were selected, chemically synthesized, and evaluated by NMR and mass spectroscopy for structural integrity. Among the ten derivatives, BPDA2 was found to be the most potent and highly selective compound, inhibiting the SHP2 enzyme activity with an IC50 of 92 nM when DiFMUP was used as a substrate and with an IC50 of 47 nM when pNPP was used as a substrate. Furthermore, enzyme kinetic analyses showed that BPDA2 is a competitive SHP2 inhibitor. Selectivity comparisons in a PTPase assay using DiFMUP as a substrate demonstrated that BPDA2 is more selective to SHP2 than to SHP1 and PTP1B by more than 369-fold and 442-fold, respectively. Evaluation with a cellular thermal shift assay (CETSA) confirmed that BPDA2 binds to wild-type SHP2 in a cellular context, and stabilizes it in solution. Treatment of cells with DBDA2 downregulates mitogenic and cell survival signaling and RTK expression in a concentration dependent manner. Furthermore, treatment of cells with BPDA2 suppresses anchorage independent growth and cancer stem cell properties of breast cancer cells. Overall, data described in this report show that BPDA2 is a more potent derivative of CNBDA with a highly improved selectivity for SHP2.
Collapse
|
12
|
Hou Y, Lu X, Xu Z, Qu J, Huang J. How a single mutation alters the protein structure: a simulation investigation on protein tyrosine phosphatase SHP2. RSC Adv 2023; 13:4263-4274. [PMID: 36760301 PMCID: PMC9891203 DOI: 10.1039/d2ra07472a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023] Open
Abstract
Protein tyrosine phosphatase SHP2 is a key regulator modulating several signaling pathways. The oncogenic mutation E76K in SHP2 releases the enzyme from an autoinhibited, closed conformation into an active, open conformation. Here, we investigated the conformational dynamics of SHP2 and the effect of the E76K mutation on its conformational ensemble via extensive molecular dynamics (MD) and metadynamics (MetaD) simulations. Our simulations provide atomistic details on how the E76K mutated SHP2 prefers the open state and also reveal that the transition between the closed and the open states is highly collective. Several intermediate metastable states during the conformational transition between the closed and the open states were also investigated. Understanding how the single E76K mutation induces the conformational change in SHP2 could facilitate the further design of SHP2 inhibitors.
Collapse
Affiliation(s)
- Yingnan Hou
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine 18 Shilongshan Road Hangzhou 310024 Zhejiang China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University 18 Shilongshan Road Hangzhou 310024 Zhejiang China
| | - Xiaoli Lu
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine 18 Shilongshan Road Hangzhou 310024 Zhejiang China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University 18 Shilongshan Road Hangzhou 310024 Zhejiang China
| | - Ziyao Xu
- BioMap 2 Kexueyuan South Road Beijing 100000 China
| | - Jiarun Qu
- BioMap 2 Kexueyuan South Road Beijing 100000 China
| | - Jing Huang
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine 18 Shilongshan Road Hangzhou 310024 Zhejiang China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University 18 Shilongshan Road Hangzhou 310024 Zhejiang China
| |
Collapse
|
13
|
Asmamaw MD, Shi XJ, Zhang LR, Liu HM. A comprehensive review of SHP2 and its role in cancer. Cell Oncol 2022; 45:729-753. [PMID: 36066752 DOI: 10.1007/s13402-022-00698-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 12/26/2022] Open
Abstract
Src homology 2-containing protein tyrosine phosphatase 2 (SHP2) is a non-receptor protein tyrosine phosphatase ubiquitously expressed mainly in the cytoplasm of several tissues. SHP2 modulates diverse cell signaling events that control metabolism, cell growth, differentiation, cell migration, transcription and oncogenic transformation. It interacts with diverse molecules in the cell, and regulates key signaling events including RAS/ERK, PI3K/AKT, JAK/STAT and PD-1 pathways downstream of several receptor tyrosine kinases (RTKs) upon stimulation by growth factors and cytokines. SHP2 acts as both a phosphatase and a scaffold, and plays prominently oncogenic functions but can be tumor suppressor in a context-dependent manner. It typically acts as a positive regulator of RTKs signaling with some inhibitory functions reported as well. SHP2 expression and activity is regulated by such factors as allosteric autoinhibition, microRNAs, ubiquitination and SUMOylation. Dysregulation of SHP2 expression or activity causes many developmental diseases, and hematological and solid tumors. Moreover, upregulated SHP2 expression or activity also decreases sensitivity of cancer cells to anticancer drugs. SHP2 is now considered as a compelling anticancer drug target and several classes of SHP2 inhibitors with different mode of action are developed with some already in clinical trial phases. Moreover, novel SHP2 substrates and functions are rapidly growing both in cell and cancer. In view of this, we comprehensively and thoroughly reviewed literatures about SHP2 regulatory mechanisms, substrates and binding partners, biological functions, roles in human cancers, and different classes of small molecule inhibitors target this oncoprotein in cancer.
Collapse
Affiliation(s)
- Moges Dessale Asmamaw
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory for Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450001, People's Republic of China
| | - Xiao-Jing Shi
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, 450052, People's Republic of China
| | - Li-Rong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory for Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan Province, 450001, People's Republic of China.
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan Province, China. .,Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, Henan Province, 450001, People's Republic of China.
| |
Collapse
|
14
|
Song Y, Yang X, Wang S, Zhao M, Yu B. Crystallographic landscape of SHP2 provides molecular insights for SHP2 targeted drug discovery. Med Res Rev 2022; 42:1781-1821. [DOI: 10.1002/med.21890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/17/2022] [Accepted: 05/04/2022] [Indexed: 12/31/2022]
Affiliation(s)
- Yihui Song
- School of Pharmaceutical Sciences Zhengzhou University 450001 Henan Zhengzhou China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment Zhengzhou University 450000 Henan Zhengzhou China
| | - Xinyu Yang
- School of Pharmaceutical Sciences Zhengzhou University 450001 Henan Zhengzhou China
| | - Shu Wang
- School of Pharmaceutical Sciences Zhengzhou University 450001 Henan Zhengzhou China
| | - Min Zhao
- School of Pharmaceutical Sciences Zhengzhou University 450001 Henan Zhengzhou China
| | - Bin Yu
- School of Pharmaceutical Sciences Zhengzhou University 450001 Henan Zhengzhou China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment Zhengzhou University 450000 Henan Zhengzhou China
| |
Collapse
|
15
|
Liu J, Qin W, Wang B, Wang Z, Hua H, Zhou F, Jia Z, Wu P, Chao H, Lu X. PTPN11 mutations in adult acute myeloid leukaemia: prevalence and clinical implications in the context of NPM1 mutation. Leuk Res 2022; 118:106859. [DOI: 10.1016/j.leukres.2022.106859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 04/14/2022] [Accepted: 05/07/2022] [Indexed: 11/30/2022]
|
16
|
Vanlallawma A, Lallawmzuali D, Pautu JL, Scaria V, Sivasubbu S, Kumar NS. Whole exome sequencing of pediatric leukemia reveals a novel InDel within FLT-3 gene in AML patient from Mizo tribal population, Northeast India. BMC Genom Data 2022; 23:23. [PMID: 35350997 PMCID: PMC8961913 DOI: 10.1186/s12863-022-01037-x] [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: 10/29/2021] [Accepted: 03/09/2022] [Indexed: 11/22/2022] Open
Abstract
Background Leukemia is the most common type of cancer in pediatrics. Genomic mutations contribute towards the molecular mechanism of disease progression and also helps in diagnosis and prognosis. This is the first scientific mutational exploration in whole exome of pediatric leukemia patients from a cancer prone endogamous Mizo tribal population, Northeast India. Result Three non-synonymous exonic variants in NOTCH1 (p.V1699E), MUTYH (p.G143E) and PTPN11 (p.S502P) were found to be pathogenic. A novel in-frame insertion-deletion within the juxtamembrane domain of FLT3 (p.Tyr589_Tyr591delinsTrpAlaGlyAsp) was also observed. Conclusion These unique variants could have a potential mutational significance and these could be candidate genes in elucidating the possibility of predisposition to cancers within the population. This study merits further investigation for its role in diagnosis and prognosis and also suggests the need for population wide screening to identify unique mutations that might play a key role towards precision medicine. Supplementary Information The online version contains supplementary material available at 10.1186/s12863-022-01037-x.
Collapse
Affiliation(s)
- Andrew Vanlallawma
- Department of Biotechnology, Mizoram University, Aizawl, Mizoram, 796004, India
| | - Doris Lallawmzuali
- Department of Pathology, Mizoram State Cancer Institute, Zemabawk, Aizawl, Mizoram, 796017, India
| | - Jeremy L Pautu
- Department of Medical Oncology, Mizoram State Cancer Institute, Zemabawk, Aizawl, Mizoram, 796017, India
| | - Vinod Scaria
- CSIR - Institute of Genomics and Integrative Biology, South Campus, Mathura Road, New Delhi, 110025, India
| | - Sridhar Sivasubbu
- CSIR - Institute of Genomics and Integrative Biology, South Campus, Mathura Road, New Delhi, 110025, India
| | | |
Collapse
|
17
|
Genomic and Epigenomic Landscape of Juvenile Myelomonocytic Leukemia. Cancers (Basel) 2022; 14:cancers14051335. [PMID: 35267643 PMCID: PMC8909150 DOI: 10.3390/cancers14051335] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myelodysplastic/myeloproliferative neoplasm characterized by the constitutive activation of the RAS pathway. In spite of the recent progresses in the molecular characterization of JMML, this disease is still a clinical challenge due to its heterogeneity, difficult diagnosis, poor prognosis, and the lack of curative treatment options other than hematopoietic stem cell transplantation (HSCT). In this review, we will provide a detailed overview of the genetic and epigenetic alterations occurring in JMML, and discuss their clinical relevance in terms of disease prognosis and risk of relapse after HSCT. We will also present the most recent advances on novel preclinical and clinical therapeutic approaches directed against JMML molecular targets. Finally, we will outline future research perspectives to further explore the oncogenic mechanism driving JMML leukemogenesis and progression, with special attention to the application of single-cell next-generation sequencing technologies. Abstract Juvenile myelomonocytic leukemia (JMML) is a rare myelodysplastic/myeloproliferative neoplasm of early childhood. Most of JMML patients experience an aggressive clinical course of the disease and require hematopoietic stem cell transplantation, which is currently the only curative treatment. JMML is characterized by RAS signaling hyperactivation, which is mainly driven by mutations in one of five genes of the RAS pathway, including PTPN11, KRAS, NRAS, NF1, and CBL. These driving mutations define different disease subtypes with specific clinico-biological features. Secondary mutations affecting other genes inside and outside the RAS pathway contribute to JMML pathogenesis and are associated with a poorer prognosis. In addition to these genetic alterations, JMML commonly presents aberrant epigenetic profiles that strongly correlate with the clinical outcome of the patients. This observation led to the recent publication of an international JMML stratification consensus, which defines three JMML clinical groups based on DNA methylation status. Although the characterization of the genomic and epigenomic landscapes in JMML has significantly contributed to better understand the molecular mechanisms driving the disease, our knowledge on JMML origin, cell identity, and intratumor and interpatient heterogeneity is still scarce. The application of new single-cell sequencing technologies will be critical to address these questions in the future.
Collapse
|
18
|
Milton CI, Selfe J, Aladowicz E, Man SYK, Bernauer C, Missiaglia E, Walters ZS, Gatz SA, Kelsey A, Generali M, Box G, Valenti M, de Haven‐Brandon A, Galiwango D, Hayes A, Clarke M, Izquierdo E, Gonzalez De Castro D, Raynaud FI, Kirkin V, Shipley JM. FGF7-FGFR2 autocrine signaling increases growth and chemoresistance of fusion-positive rhabdomyosarcomas. Mol Oncol 2022; 16:1272-1289. [PMID: 34850536 PMCID: PMC8936514 DOI: 10.1002/1878-0261.13145] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/30/2021] [Accepted: 11/29/2021] [Indexed: 11/09/2022] Open
Abstract
Rhabdomyosarcomas are aggressive pediatric soft-tissue sarcomas and include high-risk PAX3-FOXO1 fusion-gene-positive cases. Fibroblast growth factor receptor 4 (FGFR4) is known to contribute to rhabdomyosarcoma progression; here, we sought to investigate the involvement and potential for therapeutic targeting of other FGFRs in this disease. Cell-based screening of FGFR inhibitors with potential for clinical repurposing (NVP-BGJ398, nintedanib, dovitinib, and ponatinib) revealed greater sensitivity of fusion-gene-positive versus fusion-gene-negative rhabdomyosarcoma cell lines and was shown to be correlated with high expression of FGFR2 and its specific ligand, FGF7. Furthermore, patient samples exhibit higher mRNA levels of FGFR2 and FGF7 in fusion-gene-positive versus fusion-gene-negative rhabdomyosarcomas. Sustained intracellular mitogen-activated protein kinase (MAPK) activity and FGF7 secretion into culture media during serum starvation of PAX3-FOXO1 rhabdomyosarcoma cells together with decreased cell viability after genetic silencing of FGFR2 or FGF7 was in keeping with a novel FGF7-FGFR2 autocrine loop. FGFR inhibition with NVP-BGJ398 reduced viability and was synergistic with SN38, the active metabolite of irinotecan. In vivo, NVP-BGJ398 abrogated xenograft growth and warrants further investigation in combination with irinotecan as a therapeutic strategy for fusion-gene-positive rhabdomyosarcomas.
Collapse
Affiliation(s)
- Christopher I. Milton
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Signal Transduction and Molecular Pharmacology TeamCancer Research UK Cancer Therapeutics UnitThe Institute of Cancer ResearchSuttonUK
| | - Joanna Selfe
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Ewa Aladowicz
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Stella Y. K. Man
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Carolina Bernauer
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Edoardo Missiaglia
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Department of Molecular PathologyCentre Hospitalier Universitaire VaudoisLausanneSwitzerland
| | - Zoë S. Walters
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Translational Epigenomics TeamHuman Development and HealthFaculty of MedicineSouthampton General HospitalUK
| | - Susanne A. Gatz
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Institute of Cancer and Genomic SciencesUniversity of BirminghamUK
| | - Anna Kelsey
- Department of Paediatric HistopathologyManchester University NHS Foundation TrustRoyal Manchester Children’s HospitalUK
| | - Melanie Generali
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
Center for Therapy Development and Good Manufacturing PracticeInstitute for Regenerative Medicine (IREM)University of ZurichSwitzerland
| | - Gary Box
- Cancer Pharmacology and Stress Response TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Melanie Valenti
- Cancer Pharmacology and Stress Response TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Alexis de Haven‐Brandon
- Cancer Pharmacology and Stress Response TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - David Galiwango
- Drug Metabolism and Pharmacokinetics TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Angela Hayes
- Drug Metabolism and Pharmacokinetics TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Matthew Clarke
- Glioma TeamDivision of Molecular PathologyThe Institute of Cancer ResearchLondonUK
| | - Elisa Izquierdo
- Glioma TeamDivision of Molecular PathologyThe Institute of Cancer ResearchLondonUK
| | - David Gonzalez De Castro
- Molecular HaematologyDivision of Molecular PathologyThe Institute of Cancer ResearchLondonUK
- Present address:
School of MedicineDentistry and Biomedical sciencesQueens University BelfastUK
| | - Florence I. Raynaud
- Drug Metabolism and Pharmacokinetics TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Vladimir Kirkin
- Cancer Pharmacology and Stress Response TeamDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Janet M. Shipley
- Sarcoma Molecular Pathology TeamDivisions of Molecular Pathology and Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| |
Collapse
|
19
|
Song Y, Wang S, Zhao M, Yang X, Yu B. Strategies Targeting Protein Tyrosine Phosphatase SHP2 for Cancer Therapy. J Med Chem 2022; 65:3066-3079. [PMID: 35157464 DOI: 10.1021/acs.jmedchem.1c02008] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The protein tyrosine phosphatase SHP2 encoded by PTPN11 is a promising therapeutic target for cancer therapy, while the multifaceted roles of SHP2 complicate the drug discovery targeting SHP2. Given the biological significance of SHP2, strategies targeting SHP2 have been developed in recent years. To date, eight SHP2 inhibitors have advanced into clinical trials as mono- or combined therapy for treating solid tumors or adaptive resistant cancers. In this Perspective, we briefly summarize the strategies targeting SHP2 including inhibitors, activators, and proteolysis-targeting chimera (PROTAC) degraders. Besides, targeting the protein-protein interactions between SHP2 and other adaptor proteins is also discussed. Finally, we also highlight the target- and pathway-dependent combination strategies of SHP2 for cancer therapy. This Perspective may provide a timely and updated overview on the strategies targeting SHP2 for cancer therapy.
Collapse
Affiliation(s)
- Yihui Song
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China.,State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100000, China
| | - Shu Wang
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Min Zhao
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Xinyu Yang
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Yu
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China.,State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100000, China
| |
Collapse
|
20
|
Liu M, Gao S, Elhassan RM, Hou X, Fang H. Strategies to overcome drug resistance using SHP2 inhibitors. Acta Pharm Sin B 2021; 11:3908-3924. [PMID: 35024315 PMCID: PMC8727779 DOI: 10.1016/j.apsb.2021.03.037] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 03/08/2021] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
Encoded by PTPN11, the SHP2 (Src homology-2 domain-containing protein tyrosine phosphatase-2) is widely recognized as a carcinogenic phosphatase. As a promising anti-cancer drug target, SHP2 regulates many signaling pathways such as RAS-RAF-ERK, PI3K-AKT and JAK-STAT. Meanwhile, SHP2 plays a significant role in regulating immune cell function in the tumor microenvironment. Heretofore, five SHP2 allosteric inhibitors have been recruited in clinical studies for the treatment of cancer. Most recently, studies have proved the therapeutic potential of SHP2 inhibitor in overcoming drug resistance of kinase inhibitors and programmed cell death-1 (PD-1) blockade. Herein, we review the structure, function and small molecular inhibitors of SHP2, and highlight recent progress in overcoming drug resistance using SHP2 inhibitor. We hope this review would facilitate the future clinical development of SHP2 inhibitors.
Collapse
Affiliation(s)
| | | | | | - Xuben Hou
- Corresponding author. Tel./fax: +86 531 88381168.
| | - Hao Fang
- Corresponding author. Tel./fax: +86 531 88381168.
| |
Collapse
|
21
|
Calligari P, Santucci V, Stella L, Bocchinfuso G. Discriminating between competing models for the allosteric regulation of oncogenic phosphatase SHP2 by characterizing its active state. Comput Struct Biotechnol J 2021; 19:6125-6139. [PMID: 34900129 PMCID: PMC8632847 DOI: 10.1016/j.csbj.2021.10.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 11/07/2022] Open
Abstract
The Src-homology 2 domain containing phosphatase 2 (SHP2) plays a critical role in crucial signaling pathways and is involved in oncogenesis and in developmental disorders. Its structure includes two SH2 domains (N-SH2 and C-SH2), and a protein tyrosine phosphatase (PTP) domain. Under basal conditions, SHP2 is auto-inhibited, with the N-SH2 domain blocking the PTP active site. Activation involves a rearrangement of the domains that makes the catalytic site accessible, coupled to the association between the SH2 domains and cognate proteins containing phosphotyrosines. Several aspects of this transition are debated and competing mechanistic models have been proposed. A crystallographic structure of SHP2 in an active state has been reported (PDB code 6crf), but several lines of evidence suggests that it is not fully representative of the conformations populated in solution. To clarify the structural rearrangements involved in SHP2 activation, enhanced sampling simulations of the autoinhibited and active states have been performed, for wild type SHP2 and its pathogenic E76K variant. Our results demonstrate that the crystallographic conformation of the active state is unstable in solution, and multiple interdomain arrangements are populated, thus allowing association to bisphosphorylated sequences. Contrary to a recent proposal, activation is coupled to the conformational changes of the N-SH2 binding site, which is significantly more accessible in the active sate, rather than to the structure of the central β-sheet of the domain. In this coupling, a previously undescribed role for the N-SH2 BG loop emerged.
Collapse
Key Words
- BTLA, B and T lymphocyte attenuator
- CTLA-4, cytotoxic T lymphocyte-associated antigen 4
- FRET, Förster resonance energy transfer
- Inter-domain dynamics
- JMML, juvenile myelomonocytic leukemia
- MD, molecular dynamics
- NS, Noonan syndrome
- NSML, Noonan syndrome with multiple lentigines
- PD-1, programmed cell death protein 1
- PDB, protein data bank
- PMF, potential of mean force
- PTP, protein tyrosine phosphatase
- Protein flexibility
- REMD, replica exchange molecular dynamics
- RMSD, root mean square deviation
- RMSF, root mean square fluctuation
- RTK, receptor tyrosine kinase
- Replica exchange molecular dynamics simulations
- SASA, solvent accessible surface area
- SAXS, small angle X-ray scattering
- SH2, Src homology 2
- SHP2 regulatory mechanism
- SHP2, Src homology 2 domain-containing phosphatase 2
- SIRPalpha, signal regulatory protein alpha
- pY, phosphorylated tyrosine
Collapse
Affiliation(s)
- Paolo Calligari
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Valerio Santucci
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Lorenzo Stella
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| | - Gianfranco Bocchinfuso
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma Tor Vergata, Rome, Italy
| |
Collapse
|
22
|
Vemulapalli V, Donovan KA, Seegar TCM, Rogers JM, Bae M, Lumpkin RJ, Cao R, Henke MT, Ray SS, Fischer ES, Cuny GD, Blacklow SC. Targeted Degradation of the Oncogenic Phosphatase SHP2. Biochemistry 2021; 60:2593-2609. [PMID: 34411482 DOI: 10.1021/acs.biochem.1c00377] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SHP2 is a protein tyrosine phosphatase that plays a critical role in the full activation of the Ras-MAPK pathway upon stimulation of receptor tyrosine kinases, which are frequently amplified or mutationally activated in human cancer. In addition, activating mutations in SHP2 result in developmental disorders and hematologic malignancies. Several allosteric inhibitors have been developed for SHP2 and are currently in clinical trials. Here, we report the development and evaluation of a SHP2 PROTAC created by conjugating RMC-4550 with pomalidomide using a PEG linker. This molecule is highly selective for SHP2, induces degradation of SHP2 in leukemic cells at submicromolar concentrations, inhibits MAPK signaling, and suppresses cancer cell growth. SHP2 PROTACs serve as an alternative strategy for targeting ERK-dependent cancers and are useful tools alongside allosteric inhibitors for dissecting the mechanisms by which SHP2 exerts its oncogenic activity.
Collapse
Affiliation(s)
- Vidyasiri Vemulapalli
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Tom C M Seegar
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, United States
| | - Julia M Rogers
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Munhyung Bae
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ryan J Lumpkin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Ruili Cao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Matthew T Henke
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Soumya S Ray
- RA Capital, 200 Berkeley Street, Boston, Massachusetts 02116, United States
| | - Eric S Fischer
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Gregory D Cuny
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas 77204, United States
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| |
Collapse
|
23
|
Exploring the cause of the dual allosteric targeted inhibition attaching to allosteric sites enhancing SHP2 inhibition. Mol Divers 2021; 26:1567-1580. [PMID: 34338914 DOI: 10.1007/s11030-021-10286-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/22/2021] [Indexed: 10/20/2022]
Abstract
SHP2 is a protein tyrosine phosphatase (PTP) that can regulate the tyrosine phosphorylation level. Overexpression of SHP2 will promote the development of cancer diseases, so SHP2 has become one of the popular targets for the treatment of cancer. Studies have reported that both SHP099 and SHP844 are inhibitors of SHP2 and bind to different allosteric sites 1 and 2, respectively. Studies have shown that combining SHP099 with SHP844 will enhance pharmacological pathway inhibition in cells. This study uses molecular dynamic simulations to explore the dual allosteric targeted inhibition mechanism. The result shows that the residues THR108-TRP112 (allosteric site 1) move to LEU236-GLN245 (αB-αC link loop in PTP domain) , the residues of GLN79-GLN87 (allosteric site 2) get close to LEU262-GLN269 (αA-αB link loop in PTP domain) and HIS458-ARG465 (P-loop) come near to ARG501-THR507 (Q-loop) in SHP2-SHP099-SHP844 system, which makes the "inactive conformation" more stable and prevents the substrate from entering the catalytic site. Meanwhile, residue GLU110 (allosteric site 1), ARG265 (allosteric site 2), and ARG501 (Q-loop) are speculated to be the key residues that causing the SHP2 protein in auto-inhibition conformation. It is hoped that this study will provide clues for the development of the dual allosteric targeted inhibition of SHP2.
Collapse
|
24
|
Guo Y, Xu Y, Dong X, Zhang J. Cross the Undruggable Barrier, the Development of SHP2 Inhibitors: From Catalytic Site Inhibitors to Allosteric Inhibitors. ChemistrySelect 2021. [DOI: 10.1002/slct.202100186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Yu Guo
- Hangzhou Institute of Innovative Medicine College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 P.R. China
| | - Yaping Xu
- Hangzhou Institute of Innovative Medicine College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 P.R. China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 P.R. China
| | - Jianjun Zhang
- Department of Pharmacy Institution The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine) Hangzhou 310006 P.R. China
| |
Collapse
|
25
|
The loops of the N-SH2 binding cleft do not serve as allosteric switch in SHP2 activation. Proc Natl Acad Sci U S A 2021; 118:2025107118. [PMID: 33888588 DOI: 10.1073/pnas.2025107118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The Src-homology-2 domain-containing phosphatase SHP2 is a critical regulator of signal transduction, being implicated in cell growth and differentiation. Activating mutations cause developmental disorders and act as oncogenic drivers in hematologic cancers. SHP2 is activated by phosphopeptide binding to the N-SH2 domain, triggering the release of N-SH2 from the catalytic PTP domain. Based on early crystallographic data, it has been widely accepted that opening of the binding cleft of N-SH2 serves as the key "allosteric switch" driving SHP2 activation. To test the putative coupling between binding cleft opening and SHP2 activation as assumed by the allosteric switch model, we critically reviewed structural data of SHP2, and we used extensive molecular dynamics (MD) simulation and free energy calculations of isolated N-SH2 in solution, SHP2 in solution, and SHP2 in a crystal environment. Our results demonstrate that the binding cleft in N-SH2 is constitutively flexible and open in solution and that a closed cleft found in certain structures is a consequence of crystal contacts. The degree of opening of the binding cleft has only a negligible effect on the free energy of SHP2 activation. Instead, SHP2 activation is greatly favored by the opening of the central β-sheet of N-SH2. We conclude that opening of the N-SH2 binding cleft is not the key allosteric switch triggering SHP2 activation.
Collapse
|
26
|
Vemulapalli V, Chylek LA, Erickson A, Pfeiffer A, Gabriel KH, LaRochelle J, Subramanian K, Cao R, Stegmaier K, Mohseni M, LaMarche MJ, Acker MG, Sorger PK, Gygi SP, Blacklow SC. Time-resolved phosphoproteomics reveals scaffolding and catalysis-responsive patterns of SHP2-dependent signaling. eLife 2021; 10:64251. [PMID: 33755016 PMCID: PMC8024022 DOI: 10.7554/elife.64251] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/21/2021] [Indexed: 12/21/2022] Open
Abstract
SHP2 is a protein tyrosine phosphatase that normally potentiates intracellular signaling by growth factors, antigen receptors, and some cytokines, yet is frequently mutated in human cancer. Here, we examine the role of SHP2 in the responses of breast cancer cells to EGF by monitoring phosphoproteome dynamics when SHP2 is allosterically inhibited by SHP099. The dynamics of phosphotyrosine abundance at more than 400 tyrosine residues reveal six distinct response signatures following SHP099 treatment and washout. Remarkably, in addition to newly identified substrate sites on proteins such as occludin, ARHGAP35, and PLCγ2, another class of sites shows reduced phosphotyrosine abundance upon SHP2 inhibition. Sites of decreased phospho-abundance are enriched on proteins with two nearby phosphotyrosine residues, which can be directly protected from dephosphorylation by the paired SH2 domains of SHP2 itself. These findings highlight the distinct roles of the scaffolding and catalytic activities of SHP2 in effecting a transmembrane signaling response.
Collapse
Affiliation(s)
- Vidyasiri Vemulapalli
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States.,Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Lily A Chylek
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, United States
| | - Alison Erickson
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Anamarija Pfeiffer
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Khal-Hentz Gabriel
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States.,Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Jonathan LaRochelle
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States.,Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Kartik Subramanian
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, United States
| | - Ruili Cao
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States
| | - Kimberley Stegmaier
- Department of Pediatric Oncology, Dana Farber Cancer Institute, Boston, United States
| | - Morvarid Mohseni
- Novartis Institutes for Biomedical Research, Cambridge, United States
| | | | - Michael G Acker
- Novartis Institutes for Biomedical Research, Cambridge, United States
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Stephen C Blacklow
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, United States.,Department of Biological Chemistry & Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, United States
| |
Collapse
|
27
|
Tao Y, Xie J, Zhong Q, Wang Y, Zhang S, Luo F, Wen F, Xie J, Zhao J, Sun X, Long H, Ma J, Zhang Q, Long J, Fang X, Lu Y, Li D, Li M, Zhu J, Sun B, Li G, Diao J, Liu C. A novel partially open state of SHP2 points to a "multiple gear" regulation mechanism. J Biol Chem 2021; 296:100538. [PMID: 33722610 PMCID: PMC8054191 DOI: 10.1016/j.jbc.2021.100538] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 03/03/2021] [Accepted: 03/10/2021] [Indexed: 11/14/2022] Open
Abstract
The protein tyrosine phosphatase SHP2 mediates multiple signal transductions in various cellular pathways, controlled by a variety of upstream inputs. SHP2 dysregulation is causative of different types of cancers and developmental disorders, making it a promising drug target. However, how SHP2 is modulated by its different regulators remains largely unknown. Here, we use single-molecule fluorescence resonance energy transfer and molecular dynamics simulations to investigate this question. We identify a partially open, semiactive conformation of SHP2 that is intermediate between the known open and closed states. We further demonstrate a “multiple gear” regulatory mechanism, in which different activators (e.g., insulin receptor substrate-1 and CagA), oncogenic mutations (e.g., E76A), and allosteric inhibitors (e.g., SHP099) can shift the equilibrium of the three conformational states and regulate SHP2 activity to different levels. Our work reveals the essential role of the intermediate state in fine-tuning the activity of SHP2, which may provide new opportunities for drug development for relevant cancers.
Collapse
Affiliation(s)
- Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jingfei Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Qinglu Zhong
- University of the Chinese Academy of Sciences, Beijing, China; Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yongyao Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Shengnan Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Feng Luo
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Fengcai Wen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jingjing Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Jiawei Zhao
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaoou Sun
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Houfang Long
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Junfeng Ma
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qian Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xianyang Fang
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Lu
- University of the Chinese Academy of Sciences, Beijing, China; Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Li
- University of the Chinese Academy of Sciences, Beijing, China; Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Jidong Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Bo Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Guohui Li
- University of the Chinese Academy of Sciences, Beijing, China; Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China; University of the Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
28
|
Song Y, Zhao M, Wu Y, Yu B, Liu HM. A multifunctional cross-validation high-throughput screening protocol enabling the discovery of new SHP2 inhibitors. Acta Pharm Sin B 2021; 11:750-762. [PMID: 33777680 PMCID: PMC7982506 DOI: 10.1016/j.apsb.2020.10.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/01/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022] Open
Abstract
The protein tyrosine phosphatase Src homology phosphotyrosyl phosphatase 2 (SHP2) is implicated in various cancers, and targeting SHP2 has become a promising therapeutic approach. We herein described a robust cross-validation high-throughput screening protocol that combined the fluorescence-based enzyme assay and the conformation-dependent thermal shift assay for the discovery of SHP2 inhibitors. The established method can effectively exclude the false positive SHP2 inhibitors with fluorescence interference and was also successfully employed to identify new protein tyrosine phosphatase domain of SHP2 (SHP2-PTP) and allosteric inhibitors. Of note, this protocol showed potential for identifying SHP2 inhibitors against cancer-associated SHP2 mutation SHP2-E76A. After initial screening of our in-house compound library (∼2300 compounds), we identified 4 new SHP2-PTP inhibitors (0.17% hit rate) and 28 novel allosteric SHP2 inhibitors (1.22% hit rate), of which SYK-85 and WS-635 effectively inhibited SHP2-PTP (SYK-85: IC50 = 0.32 μmol/L; WS-635: IC50 = 4.13 μmol/L) and thus represent novel scaffolds for designing new SHP2-PTP inhibitors. TK-147, an allosteric inhibitor, inhibited SHP2 potently (IC50 = 0.25 μmol/L). In structure, TK-147 could be regarded as a bioisostere of the well characterized SHP2 inhibitor SHP-099, highlighting the essential structural elements for allosteric inhibition of SHP2. The principle underlying the cross-validation protocol is potentially feasible to identify allosteric inhibitors or those inactivating mutants of other proteins.
Collapse
Key Words
- AKT, protein kinase B
- ALK, anaplastic lymphoma kinase
- AML, acute myelogenous leukemia
- Allosteric inhibitors
- BTLA, B and T lymphocyte attenuator
- Bis-tris, bis-(2-hydroxyethyl)amino-tris(hydroxymethyl)methane
- DTT, dithiothreitol
- DiFMU, 6,8-difluoro-4-methylumbelliferyl hydroxid
- DiFMUP, 6,8-difluoro-4-methylumbelliferyl phosphate
- Enzyme assay
- FI, fluorescence intensity
- HEPES, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid
- HTS, high-throughput screening
- High-throughput screening
- IC50, half maximal inhibitory concentration
- JAK, janus kinase
- JMML, juvenile myelomonocytic leukaemia
- LB, lysogeny broth
- LOC, ligand only control
- LS, LEOPARD syndrome
- MAPK, mitogen-activated protein kinase
- MEK, extracellular regulated protein kinase kinases
- NPC, no protein control
- NS, Noonan syndrome
- OD, optical density
- PD-1, programmed death 1
- PI3K, phosphatidylinositol 3 kinase
- PMSF, phenylmethanesulfonyl fluoride
- PTP, protein tyrosine phosphatase
- R2, coefficient of determination
- RAS, rat sarcoma
- S/B, signal over background
- SD, standard deviation
- SDS-PAGE, sodium dodecyl sulphate polyacyrlamide gel electrophoresis
- SH2, Src homology 2
- SHP2
- SHP2, Src homology phosphotyrosyl phosphatase 2
- SHP2-PTP, protein tyrosine phosphatase domain of Src homology phosphotyrosyl phosphatase 2
- SHP2-WT, wild type Src homology phosphotyrosyl phosphatase 2
- STAT, signal transducer and activator of transcription
- Thermal shift assay
- Tm, melting temperature
- p-IRS1, phosphorylated insulin receptor substrate 1
- ΔTm, melting temperature change
Collapse
Affiliation(s)
- Yihui Song
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Min Zhao
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Yu
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
29
|
Li J, Galbo PM, Gong W, Storey AJ, Tsai YH, Yu X, Ahn JH, Guo Y, Mackintosh SG, Edmondson RD, Byrum SD, Farrar JE, He S, Cai L, Jin J, Tackett AJ, Zheng D, Wang GG. ZMYND11-MBTD1 induces leukemogenesis through hijacking NuA4/TIP60 acetyltransferase complex and a PWWP-mediated chromatin association mechanism. Nat Commun 2021; 12:1045. [PMID: 33594072 PMCID: PMC7886901 DOI: 10.1038/s41467-021-21357-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 01/22/2021] [Indexed: 12/26/2022] Open
Abstract
Recurring chromosomal translocation t(10;17)(p15;q21) present in a subset of human acute myeloid leukemia (AML) patients creates an aberrant fusion gene termed ZMYND11-MBTD1 (ZM); however, its function remains undetermined. Here, we show that ZM confers primary murine hematopoietic stem/progenitor cells indefinite self-renewal capability ex vivo and causes AML in vivo. Genomics profilings reveal that ZM directly binds to and maintains high expression of pro-leukemic genes including Hoxa, Meis1, Myb, Myc and Sox4. Mechanistically, ZM recruits the NuA4/Tip60 histone acetyltransferase complex to cis-regulatory elements, sustaining an active chromatin state enriched in histone acetylation and devoid of repressive histone marks. Systematic mutagenesis of ZM demonstrates essential requirements of Tip60 interaction and an H3K36me3-binding PWWP (Pro-Trp-Trp-Pro) domain for oncogenesis. Inhibitor of histone acetylation-'reading' bromodomain proteins, which act downstream of ZM, is efficacious in treating ZM-induced AML. Collectively, this study demonstrates AML-causing effects of ZM, examines its gene-regulatory roles, and reports an attractive mechanism-guided therapeutic strategy.
Collapse
MESH Headings
- Acetylation
- Animals
- Carcinogenesis
- Cell Cycle Proteins/chemistry
- Cell Cycle Proteins/metabolism
- Cell Differentiation
- Cell Proliferation
- Cell Transformation, Neoplastic
- Chromatin/metabolism
- Chromosomal Proteins, Non-Histone/chemistry
- Chromosomal Proteins, Non-Histone/metabolism
- Co-Repressor Proteins/chemistry
- Co-Repressor Proteins/metabolism
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/metabolism
- Disease Models, Animal
- Enhancer Elements, Genetic/genetics
- Gene Expression Regulation, Leukemic
- Genome, Human
- HEK293 Cells
- Hematopoietic Stem Cells/metabolism
- Histones/metabolism
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Lysine Acetyltransferase 5/metabolism
- Mice, Inbred BALB C
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Oncogene Proteins, Fusion/metabolism
- Protein Binding
- Protein Domains
- Transcription Factors/metabolism
- Mice
Collapse
Affiliation(s)
- Jie Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Phillip M Galbo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Weida Gong
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Aaron J Storey
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yi-Hsuan Tsai
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Xufen Yu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeong Hyun Ahn
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Yiran Guo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ricky D Edmondson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jason E Farrar
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences and Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Shenghui He
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Ling Cai
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences and Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Neurology and Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gang Greg Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
30
|
Kerr DL, Haderk F, Bivona TG. Allosteric SHP2 inhibitors in cancer: Targeting the intersection of RAS, resistance, and the immune microenvironment. Curr Opin Chem Biol 2021; 62:1-12. [PMID: 33418513 DOI: 10.1016/j.cbpa.2020.11.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/09/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023]
Abstract
The nonreceptor protein tyrosine phosphatase SHP2 (encoded by PTPN11) integrates growth and differentiation signals from receptor tyrosine kinases (RTKs) into the RAS/mitogen-activated protein kinase (MAPK) cascade. Considered 'undruggable' over three decades, SHP2 is now a potentially druggable target with the advent of allosteric SHP2 inhibitors. These agents hold promise for improving patient outcomes, showing efficacy in preclinical cancer models, where SHP2 is critical for either oncogenic signaling or resistance to current targeted agents. SHP2 inhibition may also produce immunomodulatory effects in certain tumor microenvironment cells to help cultivate antitumor immune responses. The first generation of allosteric SHP2 inhibitors is under clinical evaluation to determine safety, appropriate tolerability management, and antitumor efficacy, investigations that will dictate future clinical applications.
Collapse
Affiliation(s)
- D Lucas Kerr
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Franziska Haderk
- Department of Medicine, University of California, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| |
Collapse
|
31
|
Exploring the mechanism of the potent allosteric inhibitor compound2 on SHP2 WT and SHP2 F285S by molecular dynamics study. J Mol Graph Model 2020; 103:107807. [PMID: 33338846 DOI: 10.1016/j.jmgm.2020.107807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 11/23/2022]
Abstract
Abnormal activation of Ras/MAPK signaling pathway could trigger excessive cell division. Src-homology 2 (SH2) domain-containing protein tyrosine phosphatase (SHP2) could promote Ras/MAPK activation by integrating growth factor signals. Thus, SHP2 inhibitors had become a hot topic in the treatment of cancer. SHP2F285S, mutation in SHP2, was detected in leukemia variants. The compound 2 (3-benzyl-8-chloro-2-hydroxy-4H-benzo[4,5]thiazolo[3,2-a]pyrimidin-4-one) had been reported that it was a potent allosteric inhibitor of both SHP2 wild type (SHP2WT) and the F285S mutant (SHP2F285S). However, the mechanism of inhibition remained to be further discovered. Herein, molecular docking and molecular dynamic (MD) simulation were performed to explain the inhibition mechanism of compound 2 on SHP2WT and SHP2F285S. Overall, the molecular docking analysis revealed that compound 2 maintained the "close" structure of SHP2 protein probably by locking the C-SH2 and PTP domain. Next, post-analysis demonstrated that compound 2 might make TYR66-GLU76 of D'E-loop in N-SH2 and GLU258-LYS266 of B'C-loop, HIS458-ARG465 of P-loop, VAL497-THR507 of Q-loop in PTP domain regions tightly connect and much easier maintain "self-inhibited" conformation of SHP2F285S-compound2 than that of SHP2WT-compound2. Importantly, GLU76 of D'E-loop could play a vital role in inhibition of SHP2WT-compound2 and SHP2F285S-compound2. This work provided a reliable clue to develop novel inhibitors for leukemia related to SHP2F285S.
Collapse
|
32
|
Tripathi RKP, Ayyannan SR. Emerging chemical scaffolds with potential SHP2 phosphatase inhibitory capabilities - A comprehensive review. Chem Biol Drug Des 2020; 97:721-773. [PMID: 33191603 DOI: 10.1111/cbdd.13807] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022]
Abstract
The drug discovery panorama is cluttered with promising therapeutic targets that have been deserted because of inadequate authentication and screening failures. Molecular targets formerly tagged as "undruggable" are nowadays being more cautiously cross-examined, and whilst they stay intriguing, numerous targets are emerging more accessible. Protein tyrosine phosphatases (PTPs) excellently exemplifies a class of molecular targets that have transpired as druggable, with several small molecules and antibodies recently turned available for further development. In this respect, SHP2, a PTP, has emerged as one of the potential targets in the current pharmacological research, particularly for cancer, due to its critical role in various signalling pathways. Recently, few molecules with excellent potency have entered clinical trials, but none could reach the clinic. Consequently, search for novel, non-toxic, and specific SHP2 inhibitors are on purview. In this review, general aspects of SHP2 including its structure and mechanistic role in carcinogenesis have been presented. It also sheds light on the development of novel molecular architectures belonging to diverse chemical classes that have been proposed as SHP2-specific inhibitors along with their structure-activity relationships (SARs), stemming from chemical, mechanism-based and computer-aided studies reported since January 2015 to July 2020 (excluding patents), focusing on their potency and selectivity. The encyclopedic facts and discussions presented herein will hopefully facilitate researchers to design new ligands with better efficacy and selectivity against SHP2.
Collapse
Affiliation(s)
- Rati Kailash Prasad Tripathi
- Department of Pharmaceutical Science, Sushruta School of Medical and Paramedical Sciences, Assam University (A Central University), Silchar, Assam, India.,Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
33
|
LaMarche MJ, Acker M, Argintaru A, Bauer D, Boisclair J, Chan H, Chen CHT, Chen YN, Chen Z, Deng Z, Dore M, Dunstan D, Fan J, Fekkes P, Firestone B, Fodor M, Garcia-Fortanet J, Fortin PD, Fridrich C, Giraldes J, Glick M, Grunenfelder D, Hao HX, Hentemann M, Ho S, Jouk A, Kang ZB, Karki R, Kato M, Keen N, Koenig R, LaBonte LR, Larrow J, Liu G, Liu S, Majumdar D, Mathieu S, Meyer MJ, Mohseni M, Ntaganda R, Palermo M, Perez L, Pu M, Ramsey T, Reilly J, Sarver P, Sellers WR, Sendzik M, Shultz MD, Slisz J, Slocum K, Smith T, Spence S, Stams T, Straub C, Tamez V, Toure BB, Towler C, Wang P, Wang H, Williams SL, Yang F, Yu B, Zhang JH, Zhu S. Identification of TNO155, an Allosteric SHP2 Inhibitor for the Treatment of Cancer. J Med Chem 2020; 63:13578-13594. [PMID: 32910655 DOI: 10.1021/acs.jmedchem.0c01170] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SHP2 is a nonreceptor protein tyrosine phosphatase encoded by the PTPN11 gene and is involved in cell growth and differentiation via the MAPK signaling pathway. SHP2 also plays an important role in the programed cell death pathway (PD-1/PD-L1). As an oncoprotein as well as a potential immunomodulator, controlling SHP2 activity is of high therapeutic interest. As part of our comprehensive program targeting SHP2, we identified multiple allosteric binding modes of inhibition and optimized numerous chemical scaffolds in parallel. In this drug annotation report, we detail the identification and optimization of the pyrazine class of allosteric SHP2 inhibitors. Structure and property based drug design enabled the identification of protein-ligand interactions, potent cellular inhibition, control of physicochemical, pharmaceutical and selectivity properties, and potent in vivo antitumor activity. These studies culminated in the discovery of TNO155, (3S,4S)-8-(6-amino-5-((2-amino-3-chloropyridin-4-yl)thio)pyrazin-2-yl)-3-methyl-2-oxa-8-azaspiro[4.5]decan-4-amine (1), a highly potent, selective, orally efficacious, and first-in-class SHP2 inhibitor currently in clinical trials for cancer.
Collapse
|
34
|
Tang K, Jia YN, Yu B, Liu HM. Medicinal chemistry strategies for the development of protein tyrosine phosphatase SHP2 inhibitors and PROTAC degraders. Eur J Med Chem 2020; 204:112657. [PMID: 32738411 DOI: 10.1016/j.ejmech.2020.112657] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 12/20/2022]
Abstract
As a non-receptor protein tyrosine phosphatase encoded by the PTPN11 gene, the Src homology 2 domain-containing protein tyrosine phosphatase (SHP2) is involved in mitogen-activated protein kinase (MAPK) signaling pathway and contributes to immune surveillance via programmed cell death pathway (PD-1/PD-L1). To date, numerous SHP2 inhibitors have been developed, some of them have advanced into clinical trials. Moreover, the first PROTAC degrader SHP2-D26 has been proved to effectively induce degradation of SHP2, which may open a new avenue for targeted SHP2 therapies. In this review, we systematically summarized the development of SHP2 inhibitors with a particular focus on the structure-activity relationships (SAR) studies, crystal structures or binding models, and their modes of action.
Collapse
Affiliation(s)
- Kai Tang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Military of Education, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yao-Nan Jia
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Military of Education, Zhengzhou University, Zhengzhou, 450001, China.
| | - Bin Yu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Military of Education, Zhengzhou University, Zhengzhou, 450001, China.
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Military of Education, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
35
|
Yuan X, Bu H, Zhou J, Yang CY, Zhang H. Recent Advances of SHP2 Inhibitors in Cancer Therapy: Current Development and Clinical Application. J Med Chem 2020; 63:11368-11396. [DOI: 10.1021/acs.jmedchem.0c00249] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Xinrui Yuan
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Hong Bu
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Jinpei Zhou
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Chao-Yie Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Huibin Zhang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| |
Collapse
|
36
|
Nussinov R, Tsai C, Jang H. Autoinhibition can identify rare driver mutations and advise pharmacology. FASEB J 2019; 34:16-29. [DOI: 10.1096/fj.201901341r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/18/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section Basic Science Program Frederick National Laboratory for Cancer Research Frederick MD USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine Tel Aviv University Tel Aviv Israel
| | - Chung‐Jung Tsai
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine Tel Aviv University Tel Aviv Israel
| | - Hyunbum Jang
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine Tel Aviv University Tel Aviv Israel
| |
Collapse
|
37
|
Tani H, Kurita S, Miyamoto R, Ochiai K, Tamura K, Bonkobara M. Canine histiocytic sarcoma cell lines with SHP2 p.Glu76Gln or p.Glu76Ala mutations are sensitive to allosteric SHP2 inhibitor SHP099. Vet Comp Oncol 2019; 18:161-168. [PMID: 31339650 DOI: 10.1111/vco.12524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/24/2022]
Abstract
Some canine cases of histiocytic sarcoma (HS) carry an activating mutation in the src homology two domain-containing phosphatase 2 (SHP2) encoded by PTPN11. SHP099 is an allosteric inhibitor of SHP2 that stabilizes SHP2 in a folded, auto-inhibited conformation. Here, we examined the expression and mutation status of SHP2 in five canine HS cell lines and evaluated the growth inhibitory properties of SHP099 against these cell lines. All five of the canine HS cell lines expressed SHP2, with three of the lines each harbouring a distinct mutation in PTPN11/SHP2 (p.Glu76Gln, p.Glu76Ala and p.Gly503Val). In silico analysis suggested that p.Glu76Gln and p.Glu76Ala, but not p.Gly503Val, promote shifting of the SHP2 conformation from folded to open-active state. SHP099 potently suppressed the growth of two of the mutant cell lines (harbouring SHP2 p.Glu76Gln or p.Glu76Ala) but not that of the other three cell lines. In addition, SHP099 suppressed ERK activation in the cell line harbouring the SHP2 p.Glu76Ala mutation. The SHP2 p.Glu76Gln and p.Glu76Ala mutations are considered to be activating mutations, and the signal from SHP2 p.Glu76Ala is inferred to be transduced primarily via the ERK pathway. Moreover, SHP099-sensitive HS cells, including those with SHP2 p.Glu76Gln or p.Glu76Ala mutations, may depend on these mutations for growth. Therefore, targeting cells harbouring SHP2 p.Glu76Gln and p.Glu76Ala with SHP099 may be an approach for the treatment of canine HS.
Collapse
Affiliation(s)
- Hiroyuki Tani
- Department of Veterinary Clinical Pathology, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Sena Kurita
- Department of Veterinary Clinical Pathology, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Ryo Miyamoto
- Department of Veterinary Clinical Pathology, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Kazuhiko Ochiai
- Department of Basic Science, School of Veterinary Nursing and Technology Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Kyoichi Tamura
- Department of Veterinary Clinical Pathology, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Makoto Bonkobara
- Department of Veterinary Clinical Pathology, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| |
Collapse
|
38
|
Rehman AU, Rafiq H, Rahman MU, Li J, Liu H, Luo S, Arshad T, Wadood A, Chen HF. Gain-of-Function SHP2 E76Q Mutant Rescuing Autoinhibition Mechanism Associated with Juvenile Myelomonocytic Leukemia. J Chem Inf Model 2019; 59:3229-3239. [DOI: 10.1021/acs.jcim.9b00353] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Ashfaq Ur Rehman
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Humaira Rafiq
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Mueed Ur Rahman
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiayi Li
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Liu
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shenggan Luo
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Taaha Arshad
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Hai-Feng Chen
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
39
|
Sarver P, Acker M, Bagdanoff JT, Chen Z, Chen YN, Chan H, Firestone B, Fodor M, Fortanet J, Hao H, Hentemann M, Kato M, Koenig R, LaBonte LR, Liu G, Liu S, Liu C, McNeill E, Mohseni M, Sendzik M, Stams T, Spence S, Tamez V, Tichkule R, Towler C, Wang H, Wang P, Williams SL, Yu B, LaMarche MJ. 6-Amino-3-methylpyrimidinones as Potent, Selective, and Orally Efficacious SHP2 Inhibitors. J Med Chem 2019; 62:1793-1802. [DOI: 10.1021/acs.jmedchem.8b01726] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
40
|
Mechanism of activating mutations and allosteric drug inhibition of the phosphatase SHP2. Nat Commun 2018; 9:4507. [PMID: 30375376 PMCID: PMC6207724 DOI: 10.1038/s41467-018-06814-w] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 09/20/2018] [Indexed: 01/01/2023] Open
Abstract
Protein tyrosine phosphatase SHP2 functions as a key regulator of cell cycle control, and activating mutations cause several cancers. Here, we dissect the energy landscape of wild-type SHP2 and the oncogenic mutation E76K. NMR spectroscopy and X-ray crystallography reveal that wild-type SHP2 exchanges between closed, inactive and open, active conformations. E76K mutation shifts this equilibrium toward the open state. The previously unknown open conformation is characterized, including the active-site WPD loop in the inward and outward conformations. Binding of the allosteric inhibitor SHP099 to E76K mutant, despite much weaker, results in an identical structure as the wild-type complex. A conformational selection to the closed state reduces drug affinity which, combined with E76K’s much higher activity, demands significantly greater SHP099 concentrations to restore wild-type activity levels. The differences in structural ensembles and drug-binding kinetics of cancer-associated SHP2 forms may stimulate innovative ideas for developing more potent inhibitors for activated SHP2 mutants. The protein tyrosine phosphatase SHP2 is a key regulator of cell cycle control. Here the authors combine NMR measurements and X-ray crystallography and show that wild-type SHP2 dynamically exchanges between a closed inactive conformation and an open activated form and that the oncogenic E76K mutation shifts the equilibrium to the open state, which is reversed by binding of the allosteric inhibitor SHP099.
Collapse
|
41
|
Structural reorganization of SHP2 by oncogenic mutations and implications for oncoprotein resistance to allosteric inhibition. Nat Commun 2018; 9:4508. [PMID: 30375388 PMCID: PMC6207684 DOI: 10.1038/s41467-018-06823-9] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 09/27/2018] [Indexed: 01/18/2023] Open
Abstract
Activating mutations in PTPN11, encoding the cytosolic protein tyrosine phosphatase SHP2, result in developmental disorders and act as oncogenic drivers in patients with hematologic cancers. The allosteric inhibitor SHP099 stabilizes the wild-type SHP2 enzyme in an autoinhibited conformation that is itself destabilized by oncogenic mutations. Here, we report the impact of the highly activated and most frequently observed mutation, E76K, on the structure of SHP2, and investigate the effect of E76K and other oncogenic mutations on allosteric inhibition by SHP099. SHP2E76K adopts an open conformation but can be restored to the closed, autoinhibited conformation, near-identical to the unoccupied wild-type enzyme, when complexed with SHP099. SHP099 inhibitory activity against oncogenic SHP2 variants in vitro and in cells scales inversely with the activating strength of the mutation, indicating that either oncoselective or vastly more potent inhibitors will be necessary to suppress oncogenic signaling by the most strongly activating SHP2 mutations in cancer. Activating mutations of the non-receptor protein tyrosine phosphatase SHP2 can cause cancer. Here the authors present the crystal structure of SHP2E76K, the most frequent cancer-associated SHP2 mutation, which adopts an open-state structure and show that the allosteric inhibitor SHP099 can revert SHP2E76K to its closed, autoinhibited conformation.
Collapse
|
42
|
Hill KS, Roberts ER, Wang X, Marin E, Park TD, Son S, Ren Y, Fang B, Yoder S, Kim S, Wan L, Sarnaik AA, Koomen JM, Messina JL, Teer JK, Kim Y, Wu J, Chalfant CE, Kim M. PTPN11 Plays Oncogenic Roles and Is a Therapeutic Target for BRAF Wild-Type Melanomas. Mol Cancer Res 2018; 17:583-593. [PMID: 30355677 DOI: 10.1158/1541-7786.mcr-18-0777] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/06/2018] [Accepted: 10/15/2018] [Indexed: 02/07/2023]
Abstract
Melanoma is one of the most highly mutated cancer types. To identify functional drivers of melanoma, we searched for cross-species conserved mutations utilizing a mouse melanoma model driven by loss of PTEN and CDKN2A, and identified mutations in Kras, Erbb3, and Ptpn11. PTPN11 encodes the SHP2 protein tyrosine phosphatase that activates the RAS/RAF/MAPK pathway. Although PTPN11 is an oncogene in leukemia, lung, and breast cancers, its roles in melanoma are not clear. In this study, we found that PTPN11 is frequently activated in human melanoma specimens and cell lines and is required for full RAS/RAF/MAPK signaling activation in BRAF wild-type (either NRAS mutant or wild-type) melanoma cells. PTPN11 played oncogenic roles in melanoma by driving anchorage-independent colony formation and tumor growth. In Pten- and Cdkn2a-null mice, tet-inducible and melanocyte-specific PTPN11E76K expression significantly enhanced melanoma tumorigenesis. Melanoma cells derived from this mouse model showed doxycycline-dependent tumor growth in nude mice. Silencing PTPN11E76K expression by doxycycline withdrawal caused regression of established tumors by induction of apoptosis and senescence, and suppression of proliferation. Moreover, the PTPN11 inhibitor (SHP099) also caused regression of NRASQ61K -mutant melanoma. Using a quantitative tyrosine phosphoproteomics approach, we identified GSK3α/β as one of the key substrates that were differentially tyrosine-phosphorylated in these experiments modulating PTPN11. This study demonstrates that PTPN11 plays oncogenic roles in melanoma and regulates RAS and GSK3β signaling pathways. IMPLICATIONS: This study identifies PTPN11 as an oncogenic driver and a novel and actionable therapeutic target for BRAF wild-type melanoma.
Collapse
Affiliation(s)
- Kristen S Hill
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Evan R Roberts
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Xue Wang
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Ellen Marin
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Taeeun D Park
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Sorany Son
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Yuan Ren
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Bin Fang
- Department of Proteomics, Moffitt Cancer Center, Tampa, Florida
| | - Sean Yoder
- Molecular Genomics Core, Moffitt Cancer Center, Tampa, Florida
| | - Sungjune Kim
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida.,Department of Radiology, Moffitt Cancer Center, Tampa, Florida
| | - Lixin Wan
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Amod A Sarnaik
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - John M Koomen
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Proteomics, Moffitt Cancer Center, Tampa, Florida
| | - Jane L Messina
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Youngchul Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Jie Wu
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida.,Department of Research Service, James A. Haley Veterans Hospital, Tampa, Florida
| | - Minjung Kim
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, Florida. .,Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida.,Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida
| |
Collapse
|
43
|
Hjortness MK, Riccardi L, Hongdusit A, Zwart PH, Sankaran B, De Vivo M, Fox JM. Evolutionarily Conserved Allosteric Communication in Protein Tyrosine Phosphatases. Biochemistry 2018; 57:6443-6451. [DOI: 10.1021/acs.biochem.8b00656] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Michael K. Hjortness
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Avenue, Boulder, Colorado 80303, United States
| | - Laura Riccardi
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Akarawin Hongdusit
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Avenue, Boulder, Colorado 80303, United States
| | - Peter H. Zwart
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Marco De Vivo
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Jerome M. Fox
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Avenue, Boulder, Colorado 80303, United States
| |
Collapse
|
44
|
Hayashi T, Senda M, Suzuki N, Nishikawa H, Ben C, Tang C, Nagase L, Inoue K, Senda T, Hatakeyama M. Differential Mechanisms for SHP2 Binding and Activation Are Exploited by Geographically Distinct Helicobacter pylori CagA Oncoproteins. Cell Rep 2018; 20:2876-2890. [PMID: 28930683 DOI: 10.1016/j.celrep.2017.08.080] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/26/2017] [Accepted: 08/23/2017] [Indexed: 12/28/2022] Open
Abstract
Helicobacter pylori East Asian CagA is more closely associated with gastric cancer than Western CagA. Here we show that, upon tyrosine phosphorylation, the East Asian CagA-specific EPIYA-D segment binds to the N-SH2 domain of pro-oncogenic SHP2 phosphatase two orders of magnitude greater than Western CagA-specific EPIYA-C. This high-affinity binding is achieved via cryptic interaction between Phe at the +5 position from phosphotyrosine in EPIYA-D and a hollow on the N-SH2 phosphopeptide-binding floor. Also, duplication of EPIYA-C in Western CagA, which increases gastric cancer risk, enables divalent high-affinity binding with SHP2 via N-SH2 and C-SH2. These strong CagA bindings enforce enzymatic activation of SHP2, which endows cells with neoplastic traits. Mechanistically, N-SH2 in SHP2 is in an equilibrium between stimulatory "relaxed" and inhibitory "squeezed" states, which is fixed upon high-affinity CagA binding to the "relaxed" state that stimulates SHP2. Accordingly, East Asian CagA and Western CagA exploit distinct mechanisms for SHP2 deregulation.
Collapse
Affiliation(s)
- Takeru Hayashi
- Department of Microbiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Miki Senda
- Structural Biology Research Center, Photon Factory, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba 305-0801, Japan
| | - Nobuhiro Suzuki
- Structural Biology Research Center, Photon Factory, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba 305-0801, Japan
| | - Hiroko Nishikawa
- Department of Microbiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Max Planck-The University of Tokyo Center for Integrative Inflammology, Tokyo 113-0033, Japan
| | - Chi Ben
- Department of Microbiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Chao Tang
- Department of Microbiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Lisa Nagase
- Structural Biology Research Center, Photon Factory, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba 305-0801, Japan
| | - Kaori Inoue
- Department of Microbiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Toshiya Senda
- Structural Biology Research Center, Photon Factory, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba 305-0801, Japan; Department of Materials Structure Science, School of High Energy Accelerator Science, The Graduate University of Advanced Studies, Tsukuba 305-0801, Japan.
| | - Masanori Hatakeyama
- Department of Microbiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Max Planck-The University of Tokyo Center for Integrative Inflammology, Tokyo 113-0033, Japan.
| |
Collapse
|
45
|
Bhattacharyya S, Feferman L, Han X, Ouyang Y, Zhang F, Linhardt RJ, Tobacman JK. Decline in arylsulfatase B expression increases EGFR expression by inhibiting the protein-tyrosine phosphatase SHP2 and activating JNK in prostate cells. J Biol Chem 2018; 293:11076-11087. [PMID: 29794138 DOI: 10.1074/jbc.ra117.001244] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 05/14/2018] [Indexed: 12/24/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) has a crucial role in cell differentiation and proliferation and cancer, and its expression appears to be up-regulated when arylsulfatase B (ARSB or GalNAc-4-sulfatase) is reduced. ARSB removes 4-sulfate groups from the nonreducing end of dermatan sulfate and chondroitin 4-sulfate (C4S), and its decreased expression has previously been reported to inhibit the activity of the ubiquitous protein-tyrosine phosphatase, nonreceptor type 11 (SHP2 or PTPN11). However, the mechanism by which decline in ARSB leads to decline in SHP2 activity is unclear. Here, we show that SHP2 binds preferentially C4S, rather than chondroitin 6-sulfate, and confirm that SHP2 activity declines when ARSB is silenced. The reduction in ARSB activity, and the resultant increase in C4S, increased the expression of EGFR (Her1/ErbB1) in human prostate stem and epithelial cells. The increased expression of EGFR occurred after 1) the decline in SHP2 activity, 2) enhanced c-Jun N-terminal kinase (JNK) activity, 3) increased nuclear DNA binding by c-Jun and c-Fos, and 4) EGFR promoter activation. In response to exogenous EGF, there was increased bromodeoxyuridine incorporation, consistent with enhanced cell proliferation. These findings indicated that ARSB and chondroitin 4-sulfation affect the activation of an important dual phosphorylation threonine-tyrosine kinase and the mRNA expression of a critical tyrosine kinase receptor in prostate cells. Restoration of ARSB activity with the associated reduction in C4S may provide a new therapeutic approach for managing malignancies in which EGFR-mediated tyrosine kinase signaling pathways are active.
Collapse
Affiliation(s)
- Sumit Bhattacharyya
- From the Department of Medicine, University of Illinois and Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612 and
| | - Leo Feferman
- From the Department of Medicine, University of Illinois and Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612 and
| | - Xiaorui Han
- the Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biology and Biomedical Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Yilan Ouyang
- the Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biology and Biomedical Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Fuming Zhang
- the Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biology and Biomedical Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Robert J Linhardt
- the Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, and Biology and Biomedical Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Joanne K Tobacman
- From the Department of Medicine, University of Illinois and Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612 and
| |
Collapse
|
46
|
Hyrenius-Wittsten A, Pilheden M, Sturesson H, Hansson J, Walsh MP, Song G, Kazi JU, Liu J, Ramakrishan R, Garcia-Ruiz C, Nance S, Gupta P, Zhang J, Rönnstrand L, Hultquist A, Downing JR, Lindkvist-Petersson K, Paulsson K, Järås M, Gruber TA, Ma J, Hagström-Andersson AK. De novo activating mutations drive clonal evolution and enhance clonal fitness in KMT2A-rearranged leukemia. Nat Commun 2018; 9:1770. [PMID: 29720585 PMCID: PMC5932012 DOI: 10.1038/s41467-018-04180-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 04/11/2018] [Indexed: 02/07/2023] Open
Abstract
Activating signaling mutations are common in acute leukemia with KMT2A (previously MLL) rearrangements (KMT2A-R). These mutations are often subclonal and their biological impact remains unclear. Using a retroviral acute myeloid mouse leukemia model, we demonstrate that FLT3ITD, FLT3N676K, and NRASG12D accelerate KMT2A-MLLT3 leukemia onset. Further, also subclonal FLT3N676K mutations accelerate disease, possibly by providing stimulatory factors. Herein, we show that one such factor, MIF, promotes survival of mouse KMT2A-MLLT3 leukemia initiating cells. We identify acquired de novo mutations in Braf, Cbl, Kras, and Ptpn11 in KMT2A-MLLT3 leukemia cells that favored clonal expansion. During clonal evolution, we observe serial genetic changes at the KrasG12D locus, consistent with a strong selective advantage of additional KrasG12D. KMT2A-MLLT3 leukemias with signaling mutations enforce Myc and Myb transcriptional modules. Our results provide new insight into the biology of KMT2A-R leukemia with subclonal signaling mutations and highlight the importance of activated signaling as a contributing driver. In acute leukemia with KMT2A rearrangements (KMT2A-R), activating signaling mutations are common. Here, the authors use a retroviral acute myeloid mouse leukemia model to show that subclonal de novo activating mutations drive clonal evolution in acute leukemia with KMT2A-R and enhance clonal fitness.
Collapse
Affiliation(s)
- Axel Hyrenius-Wittsten
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
| | - Mattias Pilheden
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
| | - Helena Sturesson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
| | - Jenny Hansson
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
| | - Michael P Walsh
- Department of Pathology, St. Jude Children´s Research Hospital, Memphis, TN, 38105, USA
| | - Guangchun Song
- Department of Pathology, St. Jude Children´s Research Hospital, Memphis, TN, 38105, USA
| | - Julhash U Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 63, Lund, Sweden
| | - Jian Liu
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
| | - Ramprasad Ramakrishan
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
| | - Cristian Garcia-Ruiz
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
| | - Stephanie Nance
- Department of Oncology, St. Jude Children´s Research Hospital, Memphis, TN, 38105, USA
| | - Pankaj Gupta
- Department of Computational Biology, St. Jude Children´s Research Hospital, Memphis, TN, 38105, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children´s Research Hospital, Memphis, TN, 38105, USA
| | - Lars Rönnstrand
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 63, Lund, Sweden.,Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden.,Division of Oncology, Skane University Hospital, Lund University, 221 85, Lund, Sweden
| | - Anne Hultquist
- Department of Pathology, Skane University Hospital, Lund University, 221 85, Lund, Sweden
| | - James R Downing
- Department of Pathology, St. Jude Children´s Research Hospital, Memphis, TN, 38105, USA
| | - Karin Lindkvist-Petersson
- Medical Structural Biology, Department of Experimental Medical Science, 221 84 Lund University, Lund, Sweden
| | - Kajsa Paulsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
| | - Marcus Järås
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden
| | - Tanja A Gruber
- Department of Pathology, St. Jude Children´s Research Hospital, Memphis, TN, 38105, USA.,Department of Oncology, St. Jude Children´s Research Hospital, Memphis, TN, 38105, USA
| | - Jing Ma
- Department of Pathology, St. Jude Children´s Research Hospital, Memphis, TN, 38105, USA
| | - Anna K Hagström-Andersson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 84, Lund, Sweden.
| |
Collapse
|
47
|
Fodor M, Price E, Wang P, Lu H, Argintaru A, Chen Z, Glick M, Hao HX, Kato M, Koenig R, LaRochelle JR, Liu G, McNeill E, Majumdar D, Nishiguchi GA, Perez LB, Paris G, Quinn CM, Ramsey T, Sendzik M, Shultz MD, Williams SL, Stams T, Blacklow SC, Acker MG, LaMarche MJ. Dual Allosteric Inhibition of SHP2 Phosphatase. ACS Chem Biol 2018; 13:647-656. [PMID: 29304282 DOI: 10.1021/acschembio.7b00980] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
SHP2 is a cytoplasmic protein tyrosine phosphatase encoded by the PTPN11 gene and is involved in cell proliferation, differentiation, and survival. Recently, we reported an allosteric mechanism of inhibition that stabilizes the auto-inhibited conformation of SHP2. SHP099 (1) was identified and characterized as a moderately potent, orally bioavailable, allosteric small molecule inhibitor, which binds to a tunnel-like pocket formed by the confluence of three domains of SHP2. In this report, we describe further screening strategies that enabled the identification of a second, distinct small molecule allosteric site. SHP244 (2) was identified as a weak inhibitor of SHP2 with modest thermal stabilization of the enzyme. X-ray crystallography revealed that 2 binds and stabilizes the inactive, closed conformation of SHP2, at a distinct, previously unexplored binding site-a cleft formed at the interface of the N-terminal SH2 and PTP domains. Derivatization of 2 using structure-based design resulted in an increase in SHP2 thermal stabilization, biochemical inhibition, and subsequent MAPK pathway modulation. Downregulation of DUSP6 mRNA, a downstream MAPK pathway marker, was observed in KYSE-520 cancer cells. Remarkably, simultaneous occupation of both allosteric sites by 1 and 2 was possible, as characterized by cooperative biochemical inhibition experiments and X-ray crystallography. Combining an allosteric site 1 inhibitor with an allosteric site 2 inhibitor led to enhanced pharmacological pathway inhibition in cells. This work illustrates a rare example of dual allosteric targeted protein inhibition, demonstrates screening methodology and tactics to identify allosteric inhibitors, and enables further interrogation of SHP2 in cancer and related pathologies.
Collapse
Affiliation(s)
- Michelle Fodor
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Edmund Price
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Ping Wang
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Hengyu Lu
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Andreea Argintaru
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Zhouliang Chen
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Meir Glick
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Huai-Xiang Hao
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Mitsunori Kato
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Robert Koenig
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Jonathan R. LaRochelle
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School and Department of Cancer Biology, Dana−Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Gang Liu
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Eric McNeill
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Dyuti Majumdar
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Gisele A. Nishiguchi
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Lawrence B. Perez
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Gregory Paris
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Christopher M. Quinn
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Timothy Ramsey
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Martin Sendzik
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Michael David Shultz
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Sarah L. Williams
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Travis Stams
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Stephen C. Blacklow
- Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School and Department of Cancer Biology, Dana−Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Michael G. Acker
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Matthew J. LaMarche
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
48
|
Xie J, Si X, Gu S, Wang M, Shen J, Li H, Shen J, Li D, Fang Y, Liu C, Zhu J. Allosteric Inhibitors of SHP2 with Therapeutic Potential for Cancer Treatment. J Med Chem 2017; 60:10205-10219. [DOI: 10.1021/acs.jmedchem.7b01520] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jingjing Xie
- Interdisciplinary
Research Center on Biology and Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, 26 Qiuyue Road, Shanghai 201203, China
- University of the Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan
District, Beijing 100049, China
| | - Xiaojia Si
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer
Feld 270, 69120 Heidelberg, Germany
| | - Shoulai Gu
- Interdisciplinary
Research Center on Biology and Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, 26 Qiuyue Road, Shanghai 201203, China
| | - Mingliang Wang
- Department of Natural Products Chemistry, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Jian Shen
- Interdisciplinary
Research Center on Biology and Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, 26 Qiuyue Road, Shanghai 201203, China
- University of the Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan
District, Beijing 100049, China
| | - Haoyan Li
- Interdisciplinary
Research Center on Biology and Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, 26 Qiuyue Road, Shanghai 201203, China
- University of the Chinese Academy of Sciences, 19 A Yuquan Road, Shijingshan
District, Beijing 100049, China
| | - Jian Shen
- Viva Biotech Ltd. 334 Aidisheng Road, Shanghai 201203, China
| | - Dan Li
- Key Laboratory for
the Genetics of Developmental and Neuropsychiatric Disorders (Ministry
of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanjia Fang
- Interdisciplinary
Research Center on Biology and Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, 26 Qiuyue Road, Shanghai 201203, China
| | - Cong Liu
- Interdisciplinary
Research Center on Biology and Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, 26 Qiuyue Road, Shanghai 201203, China
| | - Jidong Zhu
- Interdisciplinary
Research Center on Biology and Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic
Chemistry, Chinese Academy of Sciences, 26 Qiuyue Road, Shanghai 201203, China
| |
Collapse
|
49
|
LaRochelle JR, Fodor M, Ellegast JM, Liu X, Vemulapalli V, Mohseni M, Stams T, Buhrlage SJ, Stegmaier K, LaMarche MJ, Acker MG, Blacklow SC. Identification of an allosteric benzothiazolopyrimidone inhibitor of the oncogenic protein tyrosine phosphatase SHP2. Bioorg Med Chem 2017; 25:6479-6485. [DOI: 10.1016/j.bmc.2017.10.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 10/18/2022]
|
50
|
Bhattacharyya S, Feferman L, Tobacman JK. Chondroitin sulfatases differentially regulate Wnt signaling in prostate stem cells through effects on SHP2, phospho-ERK1/2, and Dickkopf Wnt signaling pathway inhibitor (DKK3). Oncotarget 2017; 8:100242-100260. [PMID: 29245974 PMCID: PMC5725016 DOI: 10.18632/oncotarget.22152] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/02/2017] [Indexed: 12/20/2022] Open
Abstract
The chondroitin sulfatases N-acetylgalactosamine-4-sulfatase (ARSB) and galactosamine-N-acetyl-6-sulfatase (GALNS) remove either the 4-sulfate group at the non-reducing end of chondroitin 4-sulfate (C4S) and dermatan sulfate, or the 6-sulfate group of chondroitin 6-sulfate, chondroitin 4,6-disulfate (chondroitin sulfate E), or keratan sulfate. In human prostate cancer tissues, the ARSB activity was reduced and the GALNS activity was increased, compared to normal prostate tissue. In human prostate stem cells, when ARSB was reduced by silencing or GALNS was increased by overexpression, activity of SHP2, the ubiquitous non-receptor tyrosine phosphatase, declined, attributable to increased binding of SHP2 with C4S. This led to increases in phospho-ERK1/2, Myc/Max nuclear DNA binding, DNA methyltransferase (DNMT) activity and expression, and methylation of the Dickkopf Wnt signaling pathway inhibitor (DKK)3 promoter and to reduced DKK3 expression. Since DKK3 negatively regulates Wnt/β-catenin signaling, silencing of ARSB or overexpression of GALNS disinhibited (increased) Wnt/β-catenin signaling. These findings indicate that the chondroitin sulfatases can exert profound effects on Wnt-mediated processes, due to epigenetic effects that modulate Wnt signaling.
Collapse
Affiliation(s)
- Sumit Bhattacharyya
- Department of Medicine, Jesse Brown VA Medical Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Leo Feferman
- Department of Medicine, Jesse Brown VA Medical Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Joanne K Tobacman
- Department of Medicine, Jesse Brown VA Medical Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|