1
|
Liu S, Ma S, Liu G, Hou L, Guan Y, Liu L, Meng Y, Yu W, Liu T, Zhou L, Yuan Z, Pang S, Zhang S, Li J, Ren X, Sun Q. CK2B Induces CD8 + T-Cell Exhaustion through HDAC8-Mediated Epigenetic Reprogramming to Limit the Efficacy of Anti-PD-1 Therapy in Non-Small-Cell Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411053. [PMID: 40013761 PMCID: PMC12021095 DOI: 10.1002/advs.202411053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Anti-PD-1 therapy has left an indelible mark in the field of non-small-cell lung cancer (NSCLC) treatment; however, its efficacy is limited in clinical practice owing to differences in the degree of effector T-cell exhaustion. Casein kinase 2 (CK2) is a protein kinase that plays an important role in T-cell immunity. In this study, it is aimed to explore the potential of targeting CK2 and its regulatory subunit CK2B to prevent or reverse T-cell exhaustion, thereby enhancing the efficacy of anti-PD-1 therapy in NSCLC. In this study, it is found that CK2B expression is closely associated with T-cell exhaustion as well as the efficacy of anti-PD-1 therapy based on scRNA-seq and in vitro and in vivo experiments. Utilization of CK2 inhibitors or knockdown of CK2B expression can upregulate TBX21 expression through HDAC8-mediated epigenetic reprogramming, restoring the effector function of CD8+ T cells and enhancing the efficacy of anti-PD-1 therapy in NSCLC. These findings underscore CK2B as a promising target for overcoming the exhaustion of effector CD8+ T cells, thereby enhancing the efficacy of anti-PD-1 and adoptive cell therapies in NSCLC. Moreover, CK2B expression serves as a novel predictor of immunotherapy efficacy for NSCLC.
Collapse
Affiliation(s)
- Shaochuan Liu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of Radiation OncologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Shiya Ma
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Gen Liu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Lingjie Hou
- Department of Radiation OncologyChongqing University Cancer HospitalChongqing400030China
| | - Yong Guan
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of Radiation OncologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Liang Liu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Yuan Meng
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Wenwen Yu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Ting Liu
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Li Zhou
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Zhiyong Yuan
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of Radiation OncologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Shuju Pang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Siyuan Zhang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Junyi Li
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of Radiation OncologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| | - Qian Sun
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Prevention and TherapyTianjin300060China
- Tianjin's Clinical Research Center for CancerTianjin300060China
- Key Laboratory of Cancer Immunology and BiotherapyTianjin300060China
- Department of ImmunologyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
- Department of BiotherapyTianjin Medical University Cancer Institute and Hospital, Tianjin Medical UniversityTianjin300060China
| |
Collapse
|
2
|
Rahman MM, Tollefsbol TO. dCas9-HDAC8-EGFP fusion enables epigenetic editing of breast cancer cells by H3K9 deacetylation. Eur J Cell Biol 2024; 103:151463. [PMID: 39437453 DOI: 10.1016/j.ejcb.2024.151463] [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: 05/21/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Epigenetic editing is thriving as a robust tool for manipulating transcriptional regulation and cell fate. Despite its regulatory role in gene downregulation, epigenetic editing with histone deacetylation has been sparsely studied, especially in the context of cancer. In this current study, we have reconstructed a dCas9-HDAC8-EGFP fusion to perform histone deacetylation on the promoter of the ESR1, TERT and CDKN1C genes for the first time in breast cancer cell lines MCF-7 and MDA-MB-231 as well as in HEK293T cells. Our results demonstrated that dCas9-HDAC8-EGFP in combination with appropriate gRNAs were able to downregulate the expression of the ESR1, TERT and CDKN1C genes transcriptionally by specifically depleting the H3K9ac level on the recruitment loci. The addition of histone deacetylase inhibitors was found to neutralize the outcomes of dCas9-HDAC8-EGFP-induced epigenetic editing. Furthermore, we observed a significant downregulation of full length ERα expression in epigenetically edited MCF-7 cells with consequential alteration in cellular response toward estradiol and tamoxifen treatment due to dCas9-HDAC8-EGFP mediated epigenetic editing of the ESR1 gene. Overall, dCas9-HDAC8-EGFP is a novel circuit that enabled downregulation of crucial genes with cellular outcome in breast cancer cells by preferentially inducing H3K9 deacetylation of specific promoter regions.
Collapse
Affiliation(s)
- Mohammad Mijanur Rahman
- Department of Biology, University of Alabama at Birmingham, 902 14th Street South, Birmingham, AL 35294, USA
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 902 14th Street South, Birmingham, AL 35294, USA; Comprehensive Cancer Center, University of Alabama at Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA; Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, USA; Nutrition Obesity Research Center, University of Alabama at Birmingham, 1675 University Blvd, Birmingham, AL 35294, USA; Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294, USA.
| |
Collapse
|
3
|
Asmamaw MD, He A, Zhang LR, Liu HM, Gao Y. Histone deacetylase complexes: Structure, regulation and function. Biochim Biophys Acta Rev Cancer 2024; 1879:189150. [PMID: 38971208 DOI: 10.1016/j.bbcan.2024.189150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Histone deacetylases (HDACs) are key epigenetic regulators, and transcriptional complexes with deacetylase function are among the epigenetic corepressor complexes in the nucleus that target the epigenome. HDAC-bearing corepressor complexes such as the Sin3 complex, NuRD complex, CoREST complex, and SMRT/NCoR complex are common in biological systems. These complexes activate the otherwise inactive HDACs in a solitary state. HDAC complexes play vital roles in the regulation of key biological processes such as transcription, replication, and DNA repair. Moreover, deregulated HDAC complex function is implicated in human diseases including cancer. Therapeutic strategies targeting HDAC complexes are being sought actively. Thus, illustration of the nature and composition of HDAC complexes is vital to understanding the molecular basis of their functions under physiologic and pathologic conditions, and for designing targeted therapies. This review presents key aspects of large multiprotein HDAC-bearing complexes including their structure, function, regulatory mechanisms, implication in disease development, and role in therapeutics.
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, China
| | - Ang He
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, 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, China.
| | - Hong-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China.
| | - Ya Gao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China.
| |
Collapse
|
4
|
Khatun S, Dasgupta I, Islam R, Amin SA, Jha T, Dhaked DK, Gayen S. Unveiling critical structural features for effective HDAC8 inhibition: a comprehensive study using quantitative read-across structure-activity relationship (q-RASAR) and pharmacophore modeling. Mol Divers 2024; 28:2197-2215. [PMID: 38871969 DOI: 10.1007/s11030-024-10903-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024]
Abstract
Histone deacetylases constitute a group of enzymes that participate in several biological processes. Notably, inhibiting HDAC8 has become a therapeutic strategy for various diseases. The current inhibitors for HDAC8 lack selectivity and target multiple HDACs. Consequently, there is a growing recognition of the need for selective HDAC8 inhibitors to enhance the effectiveness of therapeutic interventions. In our current study, we have utilized a multi-faceted approach, including Quantitative Structure-Activity Relationship (QSAR) combined with Quantitative Read-Across Structure-Activity Relationship (q-RASAR) modeling, pharmacophore mapping, molecular docking, and molecular dynamics (MD) simulations. The developed q-RASAR model has a high statistical significance and predictive ability (Q2F1:0.778, Q2F2:0.775). The contributions of important descriptors are discussed in detail to gain insight into the crucial structural features in HDAC8 inhibition. The best pharmacophore hypothesis exhibits a high regression coefficient (0.969) and a low root mean square deviation (0.944), highlighting the importance of correctly orienting hydrogen bond acceptor (HBA), ring aromatic (RA), and zinc-binding group (ZBG) features in designing potent HDAC8 inhibitors. To confirm the results of q-RASAR and pharmacophore mapping, molecular docking analysis of the five potent compounds (44, 54, 82, 102, and 118) was performed to gain further insights into these structural features crucial for interaction with the HDAC8 enzyme. Lastly, MD simulation studies of the most active compound (54, mapped correctly with the pharmacophore hypothesis) and the least active compound (34, mapped poorly with the pharmacophore hypothesis) were carried out to validate the observations of the studies above. This study not only refines our understanding of essential structural features for HDAC8 inhibition but also provides a robust framework for the rational design of novel selective HDAC8 inhibitors which may offer insights to medicinal chemists and researchers engaged in the development of HDAC8-targeted therapeutics.
Collapse
Affiliation(s)
- Samima Khatun
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Indrasis Dasgupta
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Rakibul Islam
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, 700054, India
| | - Sk Abdul Amin
- Department of Pharmaceutical Technology, JIS University, 81, Nilgunj Road, Agarpara, Kolkata, West Bengal, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Devendra Kumar Dhaked
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, 700054, India
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| |
Collapse
|
5
|
Gabizon R, Tivon B, Reddi RN, van den Oetelaar MCM, Amartely H, Cossar PJ, Ottmann C, London N. A simple method for developing lysine targeted covalent protein reagents. Nat Commun 2023; 14:7933. [PMID: 38040731 PMCID: PMC10692228 DOI: 10.1038/s41467-023-42632-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/16/2023] [Indexed: 12/03/2023] Open
Abstract
Peptide-based covalent probes can target shallow protein surfaces not typically addressable using small molecules, yet there is a need for versatile approaches to convert native peptide sequences into covalent binders that can target a broad range of residues. Here we report protein-based thio-methacrylate esters-electrophiles that can be installed easily on unprotected peptides and proteins via cysteine side chains, and react efficiently and selectively with cysteine and lysine side chains on the target. Methacrylate phosphopeptides derived from 14-3-3-binding proteins irreversibly label 14-3-3σ via either lysine or cysteine residues, depending on the position of the electrophile. Methacrylate peptides targeting a conserved lysine residue exhibit pan-isoform binding of 14-3-3 proteins both in lysates and in extracellular media. Finally, we apply this approach to develop protein-based covalent binders. A methacrylate-modified variant of the colicin E9 immunity protein irreversibly binds to the E9 DNAse, resulting in significantly higher thermal stability relative to the non-covalent complex. Our approach offers a simple and versatile route to convert peptides and proteins into potent covalent binders.
Collapse
Affiliation(s)
- Ronen Gabizon
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Barr Tivon
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Rambabu N Reddi
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Maxime C M van den Oetelaar
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600MB, Eindhoven, The Netherlands
| | - Hadar Amartely
- Wolfson Centre for Applied Structural Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Peter J Cossar
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600MB, Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600MB, Eindhoven, The Netherlands
| | - Nir London
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, 7610001, Israel.
| |
Collapse
|
6
|
Toro TB, Skripnikova EV, Bornes KE, Zhang K, Watt TJ. Endogenous expression of inactive lysine deacetylases reveals deacetylation-dependent cellular mechanisms. PLoS One 2023; 18:e0291779. [PMID: 37721967 PMCID: PMC10506724 DOI: 10.1371/journal.pone.0291779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/05/2023] [Indexed: 09/20/2023] Open
Abstract
Acetylation of lysine residues is an important and common post-translational regulatory mechanism occurring on thousands of non-histone proteins. Lysine deacetylases (KDACs or HDACs) are a family of enzymes responsible for removing acetylation. To identify the biological mechanisms regulated by individual KDACs, we created HT1080 cell lines containing chromosomal point mutations, which endogenously express either KDAC6 or KDAC8 having single inactivated catalytic domain. Engineered HT1080 cells expressing inactive KDA6 or KDAC8 domains remained viable and exhibited enhanced acetylation on known substrate proteins. RNA-seq analysis revealed that many changes in gene expression were observed when KDACs were inactivated, and that these gene sets differed significantly from knockdown and knockout cell lines. Using GO ontology, we identified several critical biological processes associated specifically with catalytic activity and others attributable to non-catalytic interactions. Treatment of wild-type cells with KDAC-specific inhibitors Tubastatin A and PCI-34051 resulted in gene expression changes distinct from those of the engineered cell lines, validating this approach as a tool for evaluating in-cell inhibitor specificity and identifying off-target effects of KDAC inhibitors. Probing the functions of specific KDAC domains using these cell lines is not equivalent to doing so using previously existing methods and provides novel insight into the catalytic functions of individual KDACs by investigating the molecular and cellular changes upon genetic inactivation.
Collapse
Affiliation(s)
- Tasha B. Toro
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States of America
| | - Elena V. Skripnikova
- Division of Basic and Pharmaceutical Sciences, Xavier University of Louisiana, New Orleans, LA, United States of America
| | - Kiara E. Bornes
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States of America
| | - Kun Zhang
- Department of Computer Science, Xavier University of Louisiana, New Orleans, LA, United States of America
- Bioinformatics Core, Xavier University of Louisiana, New Orleans, LA, United States of America
| | - Terry J. Watt
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, United States of America
| |
Collapse
|
7
|
Ali M, Khramushin A, Yadav VK, Schueler-Furman O, Ivarsson Y. Elucidation of Short Linear Motif-Based Interactions of the FERM Domains of Ezrin, Radixin, Moesin, and Merlin. Biochemistry 2023. [PMID: 37224425 DOI: 10.1021/acs.biochem.3c00096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The ERM (ezrin, radixin, and moesin) family of proteins and the related protein merlin participate in scaffolding and signaling events at the cell cortex. The proteins share an N-terminal FERM [band four-point-one (4.1) ERM] domain composed of three subdomains (F1, F2, and F3) with binding sites for short linear peptide motifs. By screening the FERM domains of the ERMs and merlin against a phage library that displays peptides representing the intrinsically disordered regions of the human proteome, we identified a large number of novel ligands. We determined the affinities for the ERM and merlin FERM domains interacting with 18 peptides and validated interactions with full-length proteins through pull-down experiments. The majority of the peptides contained an apparent Yx[FILV] motif; others show alternative motifs. We defined distinct binding sites for two types of similar but distinct binding motifs (YxV and FYDF) using a combination of Rosetta FlexPepDock computational peptide docking protocols and mutational analysis. We provide a detailed molecular understanding of how the two types of peptides with distinct motifs bind to different sites on the moesin FERM phosphotyrosine binding-like subdomain and uncover interdependencies between the different types of ligands. The study expands the motif-based interactomes of the ERMs and merlin and suggests that the FERM domain acts as a switchable interaction hub.
Collapse
Affiliation(s)
- Muhammad Ali
- Department of Chemistry - BMC, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| | - Alisa Khramushin
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Vikash K Yadav
- Department of Chemistry - BMC, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ylva Ivarsson
- Department of Chemistry - BMC, Uppsala University, Husargatan 3, 751 23 Uppsala, Sweden
| |
Collapse
|
8
|
Toro TB, Bornes KE, Watt TJ. Lysine Deacetylase Substrate Selectivity: Distinct Interaction Surfaces Drive Positive and Negative Selection for Residues Following Acetyllysine. Biochemistry 2023; 62:1464-1483. [PMID: 37043688 PMCID: PMC10157890 DOI: 10.1021/acs.biochem.3c00001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Lysine acetylation is a post-translational modification that is reversed by lysine deacetylases (KDACs). The goal of this work was to identify determinants of substrate specificity for KDACs, focusing on short-range interactions occurring with residues immediately following the acetyllysine. Using a fluorescence-based in vitro assay, we determined the activity for each enzyme with a limited panel of derivative substrate peptides, revealing a distinct reactivity profile for each enzyme. We mapped the interaction surface for KDAC6, KDAC8, and KDAC1 with the +1 and +2 substrate residues (with respect to acetyllysine) based on enzyme-substrate interaction pairs observed in molecular dynamics simulations. Characteristic residues in each KDAC interact preferentially with particular substrate residues and correlate with either enhanced or inhibited activity. Although nonpolar aromatic residues generally enhanced activity with all KDACs, the manner in which each enzyme interacted with these residues is distinct. Furthermore, each KDAC has distinctive interactions that correlate with lower activity, primarily ionic in nature. KDAC8 exhibited the most diverse and widest range of effects, while KDAC6 was sensitive only to the +1 position and KDAC1 selectivity was primarily driven by negative selection. The substrate preferences were validated for KDAC6 and KDAC8 using a set of peptides derived from known acetylated proteins. Overall, we determined how KDAC6, KDAC8, and KDAC1 achieve substrate specificity with residues following the acetyllysine. These new insights into KDAC specificity will be critical for identifying novel substrates of particular KDACs, designing KDAC-specific inhibitors, and demonstrate a general framework for understanding substrate specificity for other enzyme classes.
Collapse
Affiliation(s)
- Tasha B Toro
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Kiara E Bornes
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Terry J Watt
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| |
Collapse
|
9
|
Zhou X, Chen H, Shi Y, Li J, Ma X, Du L, Hu Y, Tao M, Zhong Q, Yan D, Zhuang S, Liu N. Histone deacetylase 8 inhibition prevents the progression of peritoneal fibrosis by counteracting the epithelial-mesenchymal transition and blockade of M2 macrophage polarization. Front Immunol 2023; 14:1137332. [PMID: 36911746 PMCID: PMC9995794 DOI: 10.3389/fimmu.2023.1137332] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Background Peritoneal dialysis (PD) is an effective replacement therapy for end-stage renal disease patients. However, long-term exposure to peritoneal dialysate will lead to the development of peritoneal fibrosis. Epigenetics has been shown to play an important role in peritoneal fibrosis, but the role of histone deacetylases 8 (HDAC8) in peritoneal fibrosis have not been elucidated. In this research, we focused on the role and mechanisms of HDAC8 in peritoneal fibrosis and discussed the mechanisms involved. Methods We examined the expression of HDAC8 in the peritoneum and dialysis effluent of continuous PD patients. Then we assessed the role and mechanism of HDAC8 in peritoneal fibrosis progression in mouse model of peritoneal fibrosis induced by high glucose peritoneal dialysis fluid by using PCI-34051. In vitro, TGF-β1 or IL-4 were used to stimulate human peritoneal mesothelial cells (HPMCs) or RAW264.7 cells to establish two cell injury models to further explore the role and mechanism of HDAC8 in epithelial-mesenchymal transition (EMT) and macrophage polarization. Results We found that HDAC8 expressed highly in the peritoneum from patients with PD-related peritonitis. We further revealed that the level of HDAC8 in the dialysate increased over time, and HDAC8 was positively correlated with TGF-β1 and vascular endothelial growth factor (VEGF), and negatively correlated with cancer antigen 125. In mouse model of peritoneal fibrosis induced by high glucose dialysate, administration of PCI-34051 (a selective HDAC8 inhibitor) significantly prevented the progression of peritoneal fibrosis. Treatment with PCI-34051 blocked the phosphorylation of epidermal growth factor receptor (EGFR) and the activation of its downstream signaling pathways ERK1/2 and STAT3/HIF-1α. Inhibition of HDAC8 also reduced apoptosis. In vitro, HDAC8 silencing with PCI-34051 or siRNA inhibited TGF-β1-induced EMT and apoptosis in HPMCs. In addition, continuous high glucose dialysate or IL-4 stimulation induced M2 macrophage polarization. Blockade of HDAC8 reduced M2 macrophage polarization by inhibiting the activation of STAT6 and PI3K/Akt signaling pathways. Conclusions We demonstrated that HDAC8 promoted the EMT of HPMCs via EGFR/ERK1/2/STAT3/HIF-1α, induced M2 macrophage polarization via STAT6 and PI3K/Akt signaling pathways, and ultimately accelerated the process of peritoneal fibrosis.
Collapse
Affiliation(s)
- Xun Zhou
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Chen
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinqing Li
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyan Ma
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lin Du
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Hu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Tao
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qin Zhong
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Danying Yan
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Morse JS, Sheng YJ, Hampton JT, Sylvain LD, Das S, Alugubelli YR, Chen PC, Yang KS, Xu S, Fierke CA, Liu WR. Phage-assisted, active site-directed ligand evolution of a potent and selective histone deacetylase 8 inhibitor. Protein Sci 2022; 31:e4512. [PMID: 36382882 PMCID: PMC9703592 DOI: 10.1002/pro.4512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/04/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022]
Abstract
Phage-assisted, active site-directed ligand evolution (PADLE) is a recently developed technique that uses an amber codon-encoded noncanonical amino acid (ncAA) as an anchor to direct phage-displayed peptides to a target for an enhanced ligand identification process. 2-Amino-8-oxodecanoic acid (Aoda) is a ketone-containing ncAA residue in the macrocyclic peptide natural product apicidin that is a pan-inhibitor of Zn2+ -dependent histone deacetylases (HDACs). Its ketone serves as an anchoring point to coordinate the catalytic zinc ion in HDACs. Using a previously evolved N𝜀 -acetyl-lysyl-tRNA synthetase in combination with tRNAPyl , we showed that Aoda was efficiently incorporated into proteins in Escherichia coli by amber suppression. By propagating an amber codon-obligate phagemid library in E. coli encoding Aoda, we generated an Aoda-containing phage-displayed peptide library. Using this library to conduct PADLE against HDAC8 revealed a 7-mer peptide GH8P01F1 with Aoda-flanking amino acid residues that matched existing peptide sequences in identified HDAC8 substrates. Switching Aoda in GH8P01F1 to a more Zn2+ -chelating ncAA S-2-amino-8-hydroxyamino-8-oxooctanoic acid (Asuha) led to an extremely potent compound GH8HA01, which has an HDAC8-inhibition Ki value of 0.67 nM. GH8HA01 and its 5-mer truncation analogue Ac-GH8HA01Δ1Δ7 that has an HDAC8-inhibition Ki value of 0.31 nM are two of the most potent HDAC8 inhibitors that have been developed. Furthermore, both are highly selective against HDAC8 compared with other HDACs tested, demonstrating the great potential of using PADLE to identify highly potent and selective ligands for targets with conserved active sites among homologues.
Collapse
Affiliation(s)
- Jared S. Morse
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
| | - Yan J. Sheng
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
| | - Joshua Trae Hampton
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
| | - Lauralee D. Sylvain
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
| | - Sukant Das
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
| | - Yugendar R. Alugubelli
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
| | - Peng‐Hsun Chase Chen
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
| | - Kai S. Yang
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
| | - Shiqing Xu
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
| | - Carol A. Fierke
- Department of BiochemistryBrandeis UniversityWalthamMassachusettsUSA
| | - Wenshe Ray Liu
- Texas A&M Drug Discovery Laboratory, Department of ChemistryTexas A&M UniversityTexasUSA
- Institute of Biosciences and Technology and Department of Translational Medical Sciences, College of MedicineTexas A&M UniversityHoustonTexasUSA
- Department of Biochemistry and BiophysicsTexas A&M UniversityTexasUSA
- Department of Molecular and Cellular Medicine, College of MedicineTexas A&M UniversityTexasUSA
| |
Collapse
|
11
|
Gupta S, Azadvari N, Hosseinzadeh P. Design of Protein Segments and Peptides for Binding to Protein Targets. BIODESIGN RESEARCH 2022; 2022:9783197. [PMID: 37850124 PMCID: PMC10521657 DOI: 10.34133/2022/9783197] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/16/2022] [Indexed: 10/19/2023] Open
Abstract
Recent years have witnessed a rise in methods for accurate prediction of structure and design of novel functional proteins. Design of functional protein fragments and peptides occupy a small, albeit unique, space within the general field of protein design. While the smaller size of these peptides allows for more exhaustive computational methods, flexibility in their structure and sparsity of data compared to proteins, as well as presence of noncanonical building blocks, add additional challenges to their design. This review summarizes the current advances in the design of protein fragments and peptides for binding to targets and discusses the challenges in the field, with an eye toward future directions.
Collapse
Affiliation(s)
- Suchetana Gupta
- Knight Campus Center for Accelerating Scientific Impact, University of Oregon, Eugene OR 97403, USA
| | - Noora Azadvari
- Knight Campus Center for Accelerating Scientific Impact, University of Oregon, Eugene OR 97403, USA
| | - Parisa Hosseinzadeh
- Knight Campus Center for Accelerating Scientific Impact, University of Oregon, Eugene OR 97403, USA
| |
Collapse
|
12
|
Varga JK, Diffley K, Welker Leng KR, Fierke CA, Schueler-Furman O. Structure-based prediction of HDAC6 substrates validated by enzymatic assay reveals determinants of promiscuity and detects new potential substrates. Sci Rep 2022; 12:1788. [PMID: 35110592 PMCID: PMC8810773 DOI: 10.1038/s41598-022-05681-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/17/2022] [Indexed: 01/25/2023] Open
Abstract
Histone deacetylases play important biological roles well beyond the deacetylation of histone tails. In particular, HDAC6 is involved in multiple cellular processes such as apoptosis, cytoskeleton reorganization, and protein folding, affecting substrates such as ɑ-tubulin, Hsp90 and cortactin proteins. We have applied a biochemical enzymatic assay to measure the activity of HDAC6 on a set of candidate unlabeled peptides. These served for the calibration of a structure-based substrate prediction protocol, Rosetta FlexPepBind, previously used for the successful substrate prediction of HDAC8 and other enzymes. A proteome-wide screen of reported acetylation sites using our calibrated protocol together with the enzymatic assay provide new peptide substrates and avenues to novel potential functional regulatory roles of this promiscuous, multi-faceted enzyme. In particular, we propose novel regulatory roles of HDAC6 in tumorigenesis and cancer cell survival via the regulation of EGFR/Akt pathway activation. The calibration process and comparison of the results between HDAC6 and HDAC8 highlight structural differences that explain the established promiscuity of HDAC6.
Collapse
Affiliation(s)
- Julia K Varga
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, POB 12272, 9112102, Jerusalem, Israel
| | - Kelsey Diffley
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI, 48109, USA
| | - Katherine R Welker Leng
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI, 48109, USA
| | - Carol A Fierke
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI, 48109, USA
- Department of Biochemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Faculty of Medicine, POB 12272, 9112102, Jerusalem, Israel.
| |
Collapse
|
13
|
Toro TB, Swanier JS, Bezue JA, Broussard CG, Watt TJ. Lysine Deacetylase Substrate Selectivity: A Dynamic Ionic Interaction Specific to KDAC8. Biochemistry 2021; 60:2524-2536. [PMID: 34357750 DOI: 10.1021/acs.biochem.1c00384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lysine acetylation and deacetylation are critical for regulation of many cellular proteins. Despite the importance of this cycle, it is unclear how lysine deacetylase (KDAC) family members discriminate between acetylated proteins to react with a discrete set of substrates. Potential short-range interactions between KDAC8 and a known biologically relevant peptide substrate were identified using molecular dynamics (MD) simulations. Activity assays with a panel of peptides derived from this substrate supported a putative ionic interaction between arginine at the -1 substrate position and KDAC8 D101. Additional assays and MD simulations confirmed this novel interaction, which promotes deacetylation of substrates. Verification that a negatively charged residue at the 101 position is necessary for the ionic interaction and observed reactivity with the substrates was performed using KDAC8 derivatives. Notably, this interaction is specific to KDAC8, as KDAC1 and KDAC6 do not form this interaction and each KDAC has a different specificity profile with the peptide substrates, even though all KDACs could potentially form ionic interactions. When reacted with a panel of putative human KDAC substrates, KDAC8 preferentially deacetylated substrates containing an arginine at the -1 position. KDAC8 D101-R(-1) is a specific enzyme-substrate interaction that begins to explain how KDACs discriminate between potential substrates and how different KDAC family members can react with different subsets of acetylated proteins in cells. This multi-pronged approach will be extended to identify other critical interactions for KDAC8 substrate binding and determine critical interactions for other KDACs.
Collapse
Affiliation(s)
- Tasha B Toro
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Jordan S Swanier
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Jada A Bezue
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Christian G Broussard
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| | - Terry J Watt
- Department of Chemistry, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125-1098, United States
| |
Collapse
|
14
|
Kumar V, Kundu S, Singh A, Singh S. Understanding the role of histone deacetylase and their inhibitors in neurodegenerative disorders: Current targets and future perspective. Curr Neuropharmacol 2021; 20:158-178. [PMID: 34151764 PMCID: PMC9199543 DOI: 10.2174/1570159x19666210609160017] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/09/2021] [Accepted: 05/26/2021] [Indexed: 11/28/2022] Open
Abstract
Neurodegenerative diseases are a group of pathological conditions that cause motor inc-ordination (jerking movements), cognitive and memory impairments result from degeneration of neurons in a specific area of the brain. Oxidative stress, mitochondrial dysfunction, excitotoxicity, neuroinflammation, neurochemical imbalance and histone deacetylase enzymes (HDAC) are known to play a crucial role in neurodegeneration. HDAC is classified into four categories (class I, II, III and class IV) depending upon their location and functions. HDAC1 and 2 are involved in neurodegeneration, while HDAC3-11 and class III HDACs are beneficial as neuroprotective. HDACs are localized in different parts of the brain- HDAC1 (hippocampus and cortex), HDAC2 (nucleus), HDAC3, 4, 5, 7 and 9 (nucleus and cytoplasm), HDAC6 & HDAC7 (cytoplasm) and HDAC11 (Nucleus, cornus ammonis 1 and spinal cord). In pathological conditions, HDAC up-regulates glutamate, phosphorylation of tau, and glial fibrillary acidic proteins while down-regulating BDNF, Heat shock protein 70 and Gelsolin. Class III HDACs are divided into seven sub-classes (SIRT1-SIRT7). Sirtuins are localized in the different parts of the brain and neuron -Sirt1 (nucleus), Sirt2 (cortex, striatum, hippocampus and spinal cord), Sirt3 (mitochondria and cytoplasm), Sirt4, Sirt5 & Sirt6 (mitochondria), Sirt7 (nucleus) and Sirt8 (nucleolus). SIRTs (1, 3, 4, and 6) are involved in neuronal survival, proliferation and modulating stress response, and SIRT2 is associated with Parkinsonism, Huntington’s disease and Alzheimer’s disease, whereas SIRT6 is only associated with Alzheimer’s disease. In this critical review, we have discussed the mechanisms and therapeutic targets of HDACs that would be beneficial for the management of neurodegenerative disorders.
Collapse
Affiliation(s)
- Vishal Kumar
- Scholar, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Satyabrata Kundu
- Scholar, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Arti Singh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Shamsher Singh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
15
|
Targeted sequencing reveals the somatic mutation landscape in a Swedish breast cancer cohort. Sci Rep 2020; 10:19304. [PMID: 33168853 PMCID: PMC7653953 DOI: 10.1038/s41598-020-74580-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer (BC) is a genetically heterogeneous disease with high prevalence in Northern Europe. However, there has been no detailed investigation into the Scandinavian somatic landscape. Here, in a homogeneous Swedish cohort, we describe the somatic events underlying BC, leveraging a targeted next-generation sequencing approach. We designed a 20.5 Mb array targeting coding and regulatory regions of genes with a known role in BC (n = 765). The selected genes were either from human BC studies (n = 294) or from within canine mammary tumor associated regions (n = 471). A set of predominantly estrogen receptor positive tumors (ER + 85%) and their normal tissue counterparts (n= 61) were sequenced to ~ 140 × and 85 × mean target coverage, respectively. MuTect2 and VarScan2 were employed to detect single nucleotide variants (SNVs) and copy number aberrations (CNAs), while MutSigCV (SNVs) and GISTIC (CNAs) algorithms estimated the significance of recurrent somatic events. The significantly mutated genes (q ≤ 0.01) were PIK3CA (28% of patients), TP53 (21%) and CDH1 (11%). However, histone modifying genes contained the largest number of variants (KMT2C and ARID1A, together 28%). Mutations in KMT2C were mutually exclusive with PI3KCA mutations (p ≤ 0. 001) and half of these affect the formation of a functional PHD domain. The tumor suppressor CDK10 was deleted in 80% of the cohort while the oncogene MDM4 was amplified. Mutational signature analyses pointed towards APOBEC deaminase activity (COSMIC signature 2) and DNA mismatch repair (COSMIC signature 6). We noticed two significantly distinct patterns related to patient age; TP53 being more mutated in the younger group (29% vs 9% of patients) and CDH23 mutations were absent from the older group. The increased somatic mutation prevalence in the histone modifying genes KMT2C and ARID1A distinguishes the Swedish cohort from previous studies. KMT2C regulates enhancer activation and assists tumor proliferation in a hormone-rich environment, possibly pointing to a role in ER + BC, especially in older cases. Finally, age of onset appears to affect the mutational landscape suggesting that a larger age-diverse population incorporating more molecular subtypes should be studied to elucidate the underlying mechanisms.
Collapse
|
16
|
Jänsch N, Sugiarto WO, Muth M, Kopranovic A, Desczyk C, Ballweg M, Kirschhöfer F, Brenner‐Weiss G, Meyer‐Almes F. Switching the Switch: Ligand Induced Disulfide Formation in HDAC8. Chemistry 2020; 26:13249-13255. [PMID: 32428298 PMCID: PMC7692948 DOI: 10.1002/chem.202001712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/11/2020] [Indexed: 01/10/2023]
Abstract
Human histone deacetylase 8 is a well-recognized target for T-cell lymphoma and particularly childhood neuroblastoma. PD-404,182 was shown to be a selective covalent inhibitor of HDAC8 that forms mixed disulfides with several cysteine residues and is also able to transform thiol groups to thiocyanates. Moreover, HDAC8 was shown to be regulated by a redox switch based on the reversible formation of a disulfide bond between cysteines Cys102 and Cys153 . This study on the distinct effects of PD-404,182 on HDAC8 reveals that this compound induces the dose-dependent formation of intramolecular disulfide bridges. Therefore, the inhibition mechanism of HDAC8 by PD-404,182 involves both, covalent modification of thiols as well as ligand mediated disulfide formation. Moreover, this study provides a deep molecular insight into the regulation mechanism of HDAC8 involving several cysteines with graduated capability to form reversible disulfide bridges.
Collapse
Affiliation(s)
- Niklas Jänsch
- Department of Chemical Engineering and BiotechnologyUniversity of Applied Sciences DarmstadtStephanstraße 764295DarmstadtGermany
| | - Wisely Oki Sugiarto
- Department of Chemical Engineering and BiotechnologyUniversity of Applied Sciences DarmstadtStephanstraße 764295DarmstadtGermany
| | - Marius Muth
- Department of Chemical Engineering and BiotechnologyUniversity of Applied Sciences DarmstadtStephanstraße 764295DarmstadtGermany
- Institute of Functional Interfaces (IFG)Karlsruhe Institute of Technology (KIT)Hermann-von-Helmholtz Platz-176334Eggenstein-LeopoldshafenGermany
| | - Aleksandra Kopranovic
- Department of Chemical Engineering and BiotechnologyUniversity of Applied Sciences DarmstadtStephanstraße 764295DarmstadtGermany
| | - Charlotte Desczyk
- Department of Chemical Engineering and BiotechnologyUniversity of Applied Sciences DarmstadtStephanstraße 764295DarmstadtGermany
| | - Matthias Ballweg
- Department of Chemical Engineering and BiotechnologyUniversity of Applied Sciences DarmstadtStephanstraße 764295DarmstadtGermany
| | - Frank Kirschhöfer
- Institute of Functional Interfaces (IFG)Karlsruhe Institute of Technology (KIT)Hermann-von-Helmholtz Platz-176334Eggenstein-LeopoldshafenGermany
| | - Gerald Brenner‐Weiss
- Institute of Functional Interfaces (IFG)Karlsruhe Institute of Technology (KIT)Hermann-von-Helmholtz Platz-176334Eggenstein-LeopoldshafenGermany
| | - Franz‐Josef Meyer‐Almes
- Department of Chemical Engineering and BiotechnologyUniversity of Applied Sciences DarmstadtStephanstraße 764295DarmstadtGermany
| |
Collapse
|
17
|
Aranha MP, Jewel YSM, Beckman RA, Weiner LM, Mitchell JC, Parks JM, Smith JC. Combining Three-Dimensional Modeling with Artificial Intelligence to Increase Specificity and Precision in Peptide-MHC Binding Predictions. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:1962-1977. [PMID: 32878910 PMCID: PMC7511449 DOI: 10.4049/jimmunol.1900918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 08/01/2020] [Indexed: 02/06/2023]
Abstract
The reliable prediction of the affinity of candidate peptides for the MHC is important for predicting their potential antigenicity and thus influences medical applications, such as decisions on their inclusion in T cell-based vaccines. In this study, we present a rapid, predictive computational approach that combines a popular, sequence-based artificial neural network method, NetMHCpan 4.0, with three-dimensional structural modeling. We find that the ensembles of bound peptide conformations generated by the programs MODELLER and Rosetta FlexPepDock are less variable in geometry for strong binders than for low-affinity peptides. In tests on 1271 peptide sequences for which the experimental dissociation constants of binding to the well-characterized murine MHC allele H-2Db are known, by applying thresholds for geometric fluctuations the structure-based approach in a standalone manner drastically improves the statistical specificity, reducing the number of false positives. Furthermore, filtering candidates generated with NetMHCpan 4.0 with the structure-based predictor led to an increase in the positive predictive value (PPV) of the peptides correctly predicted to bind very strongly (i.e., K d < 100 nM) from 40 to 52% (p = 0.027). The combined method also significantly improved the PPV when tested on five human alleles, including some with limited data for training. Overall, an average increase of 10% in the PPV was found over the standalone sequence-based method. The combined method should be useful in the rapid design of effective T cell-based vaccines.
Collapse
Affiliation(s)
- Michelle P Aranha
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37916
- Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, TN 37830
| | - Yead S M Jewel
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37916
- Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, TN 37830
| | - Robert A Beckman
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC 20007
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University Medical Center, Washington, DC 20007
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057
| | - Louis M Weiner
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057
| | - Julie C Mitchell
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830
| | - Jerry M Parks
- Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, TN 37830
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830
| | - Jeremy C Smith
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37916;
- Center for Molecular Biophysics, Oak Ridge National Laboratory, Oak Ridge, TN 37830
| |
Collapse
|
18
|
Toro TB, Watt TJ. Critical review of non-histone human substrates of metal-dependent lysine deacetylases. FASEB J 2020; 34:13140-13155. [PMID: 32862458 DOI: 10.1096/fj.202001301rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/29/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022]
Abstract
Lysine acetylation is a posttranslational modification that occurs on thousands of human proteins, most of which are cytoplasmic. Acetylated proteins are involved in numerous cellular processes and human diseases. Therefore, how the acetylation/deacetylation cycle is regulated is an important question. Eleven metal-dependent lysine deacetylases (KDACs) have been identified in human cells. These enzymes, along with the sirtuins, are collectively responsible for reversing lysine acetylation. Despite several large-scale studies which have characterized the acetylome, relatively few of the specific acetylated residues have been matched to a proposed KDAC for deacetylation. To understand the function of lysine acetylation, and its association with diseases, specific KDAC-substrate pairs must be identified. Identifying specific substrates of a KDAC is complicated both by the complexity of assaying relevant activity and by the non-catalytic interactions of KDACs with cellular proteins. Here, we discuss in vitro and cell-based experimental strategies used to identify KDAC-substrate pairs and evaluate each for the purpose of directly identifying non-histone substrates of metal-dependent KDACs. We propose criteria for a combination of reproducible experimental approaches that are necessary to establish a direct enzymatic relationship. This critical analysis of the literature identifies 108 proposed non-histone substrate-KDAC pairs for which direct experimental evidence has been reported. Of these, five pairs can be considered well-established, while another thirteen pairs have both cell-based and in vitro evidence but lack independent replication and/or sufficient cell-based evidence. We present a path forward for evaluating the remaining substrate leads and reliably identifying novel KDAC substrates.
Collapse
Affiliation(s)
- Tasha B Toro
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| | - Terry J Watt
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| |
Collapse
|
19
|
Khramushin A, Marcu O, Alam N, Shimony O, Padhorny D, Brini E, Dill KA, Vajda S, Kozakov D, Schueler-Furman O. Modeling beta-sheet peptide-protein interactions: Rosetta FlexPepDock in CAPRI rounds 38-45. Proteins 2020; 88:1037-1049. [PMID: 31891416 PMCID: PMC7539656 DOI: 10.1002/prot.25871] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 01/09/2023]
Abstract
Peptide-protein docking is challenging due to the considerable conformational freedom of the peptide. CAPRI rounds 38-45 included two peptide-protein interactions, both characterized by a peptide forming an additional beta strand of a beta sheet in the receptor. Using the Rosetta FlexPepDock peptide docking protocol we generated top-performing, high-accuracy models for targets 134 and 135, involving an interaction between a peptide derived from L-MAG with DLC8. In addition, we were able to generate the only medium-accuracy models for a particularly challenging target, T121. In contrast to the classical peptide-mediated interaction, in which receptor side chains contact both peptide backbone and side chains, beta-sheet complementation involves a major contribution to binding by hydrogen bonds between main chain atoms. To establish how binding affinity and specificity are established in this special class of peptide-protein interactions, we extracted PeptiDBeta, a benchmark of solved structures of different protein domains that are bound by peptides via beta-sheet complementation, and tested our protocol for global peptide-docking PIPER-FlexPepDock on this dataset. We find that the beta-strand part of the peptide is sufficient to generate approximate and even high resolution models of many interactions, but inclusion of adjacent motif residues often provides additional information necessary to achieve high resolution model quality.
Collapse
Affiliation(s)
- Alisa Khramushin
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| | - Orly Marcu
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| | - Nawsad Alam
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| | - Orly Shimony
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| | - Dzmitry Padhorny
- Department of Applied Mathematics and Statistics, Stony
Brook University, New York, New York
- Laufer Center for Physical and Quantitative Biology, Stony
Brook University, New York, New York
| | - Emiliano Brini
- Laufer Center for Physical and Quantitative Biology, Stony
Brook University, New York, New York
| | - Ken A. Dill
- Laufer Center for Physical and Quantitative Biology, Stony
Brook University, New York, New York
- Department of Physics and Astronomy, Stony Brook
University, New York, New York
- Department of Chemistry, Stony Brook University, New York,
New York
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University,
Boston, Massachusetts
- Department of Chemistry, Boston University, Boston,
Massachusetts
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony
Brook University, New York, New York
- Laufer Center for Physical and Quantitative Biology, Stony
Brook University, New York, New York
| | - Ora Schueler-Furman
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| |
Collapse
|
20
|
Saccoccia F, Brindisi M, Gimmelli R, Relitti N, Guidi A, Saraswati AP, Cavella C, Brogi S, Chemi G, Butini S, Papoff G, Senger J, Herp D, Jung M, Campiani G, Gemma S, Ruberti G. Screening and Phenotypical Characterization of Schistosoma mansoni Histone Deacetylase 8 ( SmHDAC8) Inhibitors as Multistage Antischistosomal Agents. ACS Infect Dis 2020; 6:100-113. [PMID: 31661956 DOI: 10.1021/acsinfecdis.9b00224] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Schistosomiasis (also known as bilharzia) is a neglected tropical disease caused by platyhelminths of the genus Schistosoma. The disease is endemic in tropical and subtropical areas of the world where water is infested by the intermediate parasite host, the snail. More than 800 million people live in endemic areas and more than 200 million are infected and require treatment. Praziquantel (PZQ) is the drug of choice for schistosomiasis treatment and transmission control being safe and very effective against adult worms of all the clinically relevant Schistosoma species. Unfortunately, it is ineffective on immature, juvenile worms; therefore, it does not prevent reinfection. Moreover, the risk of development and spread of drug resistance because of the widespread use of a single drug in such a large population represents a serious threat. Therefore, research aimed at identifying novel drugs to be used alone or in combination with PZQ are needed. Schistosoma mansoni histone deacetylase 8 (SmHDAC8) is a class I zinc-dependent HDAC, which is abundantly expressed in all stages of its life cycle, thus representing an interesting target for drug discovery. Through virtual screening and phenotypical characterization of selected hits, we discovered two main chemical classes of compounds characterized by the presence of a hydroxamate-based metal binding group coupled to a spiroindoline or a tricyclic thieno[3,2-b]indole core as capping groups. Some of the compounds of both classes were deeply investigated and showed to impair viability of larval, juvenile, and adult schistosomes, to impact egg production in vitro and/or to induce morphological alterations of the adult schistosome reproductive systems. Noteworthy, all of them inhibit the recombinant form of SmHDAC8 enzyme in vitro. Overall, we identified very interesting scaffolds, paving the way to the development of effective antischistosomal agents.
Collapse
Affiliation(s)
- Fulvio Saccoccia
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Campus A. Buzzati-Traverso, via E. Ramarini 32, 00015 Monterotondo (Rome), Italy
| | - Margherita Brindisi
- Department of Excellence of Pharmacy, University of Napoli Federico II, via D. Montesano 49, 80131 Naples, Italy
| | - Roberto Gimmelli
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Campus A. Buzzati-Traverso, via E. Ramarini 32, 00015 Monterotondo (Rome), Italy
| | - Nicola Relitti
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Alessandra Guidi
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Campus A. Buzzati-Traverso, via E. Ramarini 32, 00015 Monterotondo (Rome), Italy
| | - A Prasanth Saraswati
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Caterina Cavella
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Simone Brogi
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126, Pisa, Italy
| | - Giulia Chemi
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Stefania Butini
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Giuliana Papoff
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Campus A. Buzzati-Traverso, via E. Ramarini 32, 00015 Monterotondo (Rome), Italy
| | - Johanna Senger
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Daniel Herp
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Giuseppe Campiani
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Sandra Gemma
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Giovina Ruberti
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Campus A. Buzzati-Traverso, via E. Ramarini 32, 00015 Monterotondo (Rome), Italy
| |
Collapse
|
21
|
Leng KRW, Castañeda CA, Decroos C, Islam B, Haider SM, Christianson DW, Fierke CA. Phosphorylation of Histone Deacetylase 8: Structural and Mechanistic Analysis of the Phosphomimetic S39E Mutant. Biochemistry 2019; 58:4480-4493. [PMID: 31633931 PMCID: PMC6903415 DOI: 10.1021/acs.biochem.9b00653] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Histone deacetylase (HDAC) enzymes that catalyze removal of acetyl-lysine post-translational modifications are frequently post-translationally modified. HDAC8 is phosphorylated within the deacetylase domain at conserved residue serine 39, which leads to decreased catalytic activity. HDAC8 phosphorylation at S39 is unique in its location and function and may represent a novel mode of deacetylation regulation. To better understand the impact of phosphorylation of HDAC8 on enzyme structure and function, we performed crystallographic, kinetic, and molecular dynamics studies of the S39E HDAC8 phosphomimetic mutant. This mutation decreases the level of deacetylation of peptides derived from acetylated nuclear and cytoplasmic proteins. However, the magnitude of the effect depends on the peptide sequence and the identity of the active site metal ion [Zn(II) vs Fe(II)], with the value of kcat/KM for the mutant decreasing 9- to >200-fold compared to that of wild-type HDAC8. Furthermore, the dissociation rate constant of the active site metal ion increases by ∼10-fold. S39E HDAC8 was crystallized in complex with the inhibitor Droxinostat, revealing that phosphorylation of S39, as mimicked by the glutamate side chain, perturbs local structure through distortion of the L1 loop. Molecular dynamics simulations of both S39E and phosphorylated S39 HDAC8 demonstrate that the perturbation of the L1 loop likely occurs because of the lost hydrogen bond between D29 and S39. Furthermore, the S39 perturbation causes structural changes that propagate through the protein scaffolding to influence function in the active site. These data demonstrate that phosphorylation plays an important regulatory role for HDAC8 by affecting ligand binding, catalytic efficiency, and substrate selectivity.
Collapse
Affiliation(s)
| | - Carol Ann Castañeda
- Interdepartmental Program in Chemical Biology, University of Michigan, 210 Washtenaw Avenue 4008 Life Sciences Institute, Ann Arbor, MI 48109
| | - Christophe Decroos
- Department of Chemistry, University of Pennsylvania, 231 S. 34 Street, Philadelphia, PA 19104
| | - Barira Islam
- School of Pharmacy, University College London, 29-39 Brunswick Square London, WC1N 1AX, UK
| | - Shozeb M. Haider
- School of Pharmacy, University College London, 29-39 Brunswick Square London, WC1N 1AX, UK
| | - David W. Christianson
- Department of Chemistry, University of Pennsylvania, 231 S. 34 Street, Philadelphia, PA 19104
| | - Carol A. Fierke
- Department of Chemistry, University of Michigan, 930 N. University Ave., Ann Arbor, MI 48109
- Interdepartmental Program in Chemical Biology, University of Michigan, 210 Washtenaw Avenue 4008 Life Sciences Institute, Ann Arbor, MI 48109
- Department of Chemistry, Texas A&M University, Jack K. Williams Administration Building, Suite 100 College Station, TX 77843
| |
Collapse
|
22
|
Sequential, Structural and Functional Properties of Protein Complexes Are Defined by How Folding and Binding Intertwine. J Mol Biol 2019; 431:4408-4428. [DOI: 10.1016/j.jmb.2019.07.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/10/2019] [Accepted: 07/29/2019] [Indexed: 12/15/2022]
|
23
|
Gong D, Zeng Z, Yi F, Wu J. Inhibition of histone deacetylase 11 promotes human liver cancer cell apoptosis. Am J Transl Res 2019; 11:983-990. [PMID: 30899397 PMCID: PMC6413277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/21/2018] [Indexed: 06/09/2023]
Abstract
The apoptosis machinery is compromised in liver cancer (LC). The underlying mechanism needs to be further investigated. Histone deacetylases (HDAC) have multiple and strong biochemical activities. This study tests a hypothesis that HDAC11 prevents LC cell (LCC) apoptosis via modulating the p53 gene transcription. In this study, the LC tissues were collected from patients with LC. The LCCs were purified by magnetic cell sorting. The gene transcription activities of the LCCs were analyzed by immunoprecipitation (IP) and chromatin IP. We observed that the LCCs expressed high levels of HDAC11, which was negatively correlated with the expression of p53 in LCCs. Further findings indicated that HDAC11 formed a complex with Egr1, the transcription factor of p53. HDAC11 induced Egr1 deacetylation and thus prevented the p53 gene transcription. Over expression of HDAC11 in liver cells inhibited the cell apoptosis. Inhibition of the expression of HDAC11 in LCCs promoted the LCC apoptosis. In conclusion, HDAC11 plays a critical role in the compromising the expression p53 in LCC, which can be reversed by the inhibition of HDAC11. To regulate HDAC11 may have therapeutic potential for LC treatment.
Collapse
Affiliation(s)
- Dan Gong
- Department of Oncology, Second Affiliated Hospital of Nanchang UniversityNanchang 330006, China
| | - Zhiwen Zeng
- Department of Oncology, Third Hospital of NanchangNanchang 330009, China
| | - Fengming Yi
- Department of Oncology, Second Affiliated Hospital of Nanchang UniversityNanchang 330006, China
| | - Jianbing Wu
- Department of Oncology, Second Affiliated Hospital of Nanchang UniversityNanchang 330006, China
| |
Collapse
|
24
|
Engel JA, Norris EL, Gilson P, Przyborski J, Shonhai A, Blatch GL, Skinner-Adams TS, Gorman J, Headlam M, Andrews KT. Proteomic analysis of Plasmodium falciparum histone deacetylase 1 complex proteins. Exp Parasitol 2019; 198:7-16. [PMID: 30682336 DOI: 10.1016/j.exppara.2019.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/01/2018] [Accepted: 01/20/2019] [Indexed: 01/12/2023]
Abstract
Plasmodium falciparum histone deacetylases (PfHDACs) are an important class of epigenetic regulators that alter protein lysine acetylation, contributing to regulation of gene expression and normal parasite growth and development. PfHDACs are therefore under investigation as drug targets for malaria. Despite this, our understanding of the biological roles of these enzymes is only just beginning to emerge. In higher eukaryotes, HDACs function as part of multi-protein complexes and act on both histone and non-histone substrates. Here, we present a proteomics analysis of PfHDAC1 immunoprecipitates, identifying 26 putative P. falciparum complex proteins in trophozoite-stage asexual intraerythrocytic parasites. The co-migration of two of these (P. falciparum heat shock proteins 70-1 and 90) with PfHDAC1 was validated using Blue Native PAGE combined with Western blot. These data provide a snapshot of possible PfHDAC1 interactions and a starting point for future studies focused on elucidating the broader function of PfHDACs in Plasmodium parasites.
Collapse
Affiliation(s)
- Jessica A Engel
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Emma L Norris
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Paul Gilson
- Burnet Institute, Monash University, Victoria, Australia
| | - Jude Przyborski
- Centre of Infectious Diseases, Parasitology, University Hospital Heidelberg, Germany
| | - Addmore Shonhai
- Biochemistry Department, University of Venda, Thohoyandou, South Africa
| | - Gregory L Blatch
- The Vice Chancellery, The University of Notre Dame Australia, Fremantle, WA, Australia
| | - Tina S Skinner-Adams
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Jeffrey Gorman
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | | | - Katherine T Andrews
- Griffith Institute for Drug Discovery, Griffith University, Queensland, Australia.
| |
Collapse
|
25
|
Ochoa R, Soler MA, Laio A, Cossio P. Assessing the capability of in silico mutation protocols for predicting the finite temperature conformation of amino acids. Phys Chem Chem Phys 2018; 20:25901-25909. [PMID: 30289133 DOI: 10.1039/c8cp03826k] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mutation protocols are a key tool in computational biophysics for modelling unknown side chain conformations. In particular, these protocols are used to generate the starting structures for molecular dynamics simulations. The accuracy of the initial side chain and backbone placement is crucial to obtain a stable and quickly converging simulation. In this work, we assessed the performance of several mutation protocols in predicting the most probable conformer observed in finite temperature molecular dynamics simulations for a set of protein-peptide crystals differing only by single-point mutations in the peptide sequence. Our results show that several programs which predict well the crystal conformations fail to predict the most probable finite temperature configuration. Methods relying on backbone-dependent rotamer libraries have, in general, a better performance, but even the best protocol fails in predicting approximately 30% of the mutations.
Collapse
Affiliation(s)
- Rodrigo Ochoa
- Biophysics of Tropical Diseases, Max Planck Tandem Group, University of Antioquia, Medellin, Colombia.
| | | | | | | |
Collapse
|
26
|
Hánová I, Brynda J, Houštecká R, Alam N, Sojka D, Kopáček P, Marešová L, Vondrášek J, Horn M, Schueler-Furman O, Mareš M. Novel Structural Mechanism of Allosteric Regulation of Aspartic Peptidases via an Evolutionarily Conserved Exosite. Cell Chem Biol 2018; 25:318-329.e4. [PMID: 29396291 DOI: 10.1016/j.chembiol.2018.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/04/2017] [Accepted: 12/28/2017] [Indexed: 11/25/2022]
Abstract
Pepsin-family aspartic peptidases are biosynthesized as inactive zymogens in which the propeptide blocks the active site until its proteolytic removal upon enzyme activation. Here, we describe a novel dual regulatory function for the propeptide using a set of crystal structures of the parasite cathepsin D IrCD1. In the IrCD1 zymogen, intramolecular autoinhibition by the intact propeptide is mediated by an evolutionarily conserved exosite on the enzyme core. After activation, the mature enzyme employs the same exosite to rebind a small fragment derived from the cleaved propeptide. This fragment functions as an effective natural inhibitor of mature IrCD1 that operates in a pH-dependent manner through a unique allosteric inhibition mechanism. The study uncovers the propeptide-binding exosite as a target for the regulation of pepsin-family aspartic peptidases and defines the structural requirements for exosite inhibition.
Collapse
Affiliation(s)
- Iva Hánová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610 Prague, Czech Republic; Department of Biochemistry, Faculty of Science, Charles University, 12840 Prague, Czech Republic
| | - Jiří Brynda
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610 Prague, Czech Republic
| | - Radka Houštecká
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610 Prague, Czech Republic; First Faculty of Medicine, Charles University, 12108 Prague, Czech Republic
| | - Nawsad Alam
- Department of Microbiology and Molecular Genetics, Institute for Biomedical Research IMRIC, Hebrew University, Hadassah Medical School, 91120 Jerusalem, Israel
| | - Daniel Sojka
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| | - Petr Kopáček
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| | - Lucie Marešová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610 Prague, Czech Republic
| | - Jiří Vondrášek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610 Prague, Czech Republic
| | - Martin Horn
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610 Prague, Czech Republic
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Biomedical Research IMRIC, Hebrew University, Hadassah Medical School, 91120 Jerusalem, Israel
| | - Michael Mareš
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610 Prague, Czech Republic.
| |
Collapse
|
27
|
Castañeda CA, Wolfson NA, Leng KR, Kuo YM, Andrews AJ, Fierke CA. HDAC8 substrate selectivity is determined by long- and short-range interactions leading to enhanced reactivity for full-length histone substrates compared with peptides. J Biol Chem 2017; 292:21568-21577. [PMID: 29109148 PMCID: PMC5766737 DOI: 10.1074/jbc.m117.811026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/05/2017] [Indexed: 01/03/2023] Open
Abstract
Histone deacetylases (HDACs) catalyze deacetylation of acetyl-lysine residues within proteins. To date, HDAC substrate specificity and selectivity have been largely estimated using peptide substrates. However, it is unclear whether peptide substrates accurately reflect the substrate selectivity of HDAC8 toward full-length proteins. Here, we compare HDAC8 substrate selectivity in the context of peptides, full-length proteins, and protein-nucleic acid complexes. We demonstrate that HDAC8 catalyzes deacetylation of tetrameric histone (H3/H4) substrates with catalytic efficiencies that are 40-300-fold higher than those for corresponding peptide substrates. Thus, we conclude that additional contacts with protein substrates enhance catalytic efficiency. However, the catalytic efficiency decreases for larger multiprotein complexes. These differences in HDAC8 substrate selectivity for peptides and full-length proteins suggest that HDAC8 substrate preference is based on a combination of short- and long-range interactions. In summary, this work presents detailed kinetics for HDAC8-catalyzed deacetylation of singly-acetylated, full-length protein substrates, revealing that HDAC8 substrate selectivity is determined by multiple factors. These insights provide a foundation for understanding recognition of full-length proteins by HDACs.
Collapse
Affiliation(s)
| | | | - Katherine R Leng
- Chemistry, University of Michigan, Ann Arbor, Michigan 48109 and
| | - Yin-Ming Kuo
- the Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | | | - Carol A Fierke
- From the Program in Chemical Biology and
- the Departments of Biological Chemistry and
- Chemistry, University of Michigan, Ann Arbor, Michigan 48109 and
| |
Collapse
|
28
|
Caby S, Pagliazzo L, Lancelot J, Saliou JM, Bertheaume N, Pierce RJ, Roger E. Analysis of the interactome of Schistosoma mansoni histone deacetylase 8. PLoS Negl Trop Dis 2017; 11:e0006089. [PMID: 29155817 PMCID: PMC5722368 DOI: 10.1371/journal.pntd.0006089] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 12/08/2017] [Accepted: 10/31/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Histone deacetylase 8 from Schistosoma mansoni (SmHDAC8) is essential to parasite growth and development within the mammalian host and is under investigation as a target for the development of selective inhibitors as novel schistosomicidal drugs. Although some protein substrates and protein partners of human HDAC8 have been characterized, notably indicating a role in the function of the cohesin complex, nothing is known of the partners and biological function of SmHDAC8. METHODOLOGY/PRINCIPAL FINDINGS We therefore employed two strategies to characterize the SmHDAC8 interactome. We first used SmHDAC8 as a bait protein in yeast two-hybrid (Y2H) screening of an S. mansoni cDNA library. This allowed the identification of 49 different sequences encoding proteins. We next performed co-immunoprecipitation (Co-IP) experiments on parasite extracts with an anti-SmHDAC8 antibody. Mass spectrometry (MS) analysis allowed the identification of 160 different proteins. CONCLUSIONS/SIGNIFICANCE SmHDAC8 partners are involved in about 40 different processes, included expected functions such as the cohesin complex, cytoskeleton organization, transcriptional and translational regulation, metabolism, DNA repair, the cell cycle, protein dephosphorylation, proteolysis, protein transport, but also some proteasome and ribosome components were detected. Our results show that SmHDAC8 is a versatile deacetylase, potentially involved in both cytosolic and nuclear processes.
Collapse
Affiliation(s)
- Stéphanie Caby
- Univ. Lille, CNRS UMR 8204, INSERM U1019, CHU Lille, Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille (CIIL), F-59000 Lille, France
| | - Lucile Pagliazzo
- Univ. Lille, CNRS UMR 8204, INSERM U1019, CHU Lille, Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille (CIIL), F-59000 Lille, France
| | - Julien Lancelot
- Univ. Lille, CNRS UMR 8204, INSERM U1019, CHU Lille, Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille (CIIL), F-59000 Lille, France
| | - Jean-Michel Saliou
- Univ. Lille, CNRS UMR 8204, INSERM U1019, CHU Lille, Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille (CIIL), F-59000 Lille, France
| | - Nicolas Bertheaume
- Univ. Lille, CNRS UMR 8204, INSERM U1019, CHU Lille, Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille (CIIL), F-59000 Lille, France
| | - Raymond J. Pierce
- Univ. Lille, CNRS UMR 8204, INSERM U1019, CHU Lille, Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille (CIIL), F-59000 Lille, France
| | - Emmanuel Roger
- Univ. Lille, CNRS UMR 8204, INSERM U1019, CHU Lille, Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille (CIIL), F-59000 Lille, France
- * E-mail:
| |
Collapse
|
29
|
Lopez JE, Haynes SE, Majmudar JD, Martin BR, Fierke CA. HDAC8 Substrates Identified by Genetically Encoded Active Site Photocrosslinking. J Am Chem Soc 2017; 139:16222-16227. [PMID: 29035536 DOI: 10.1021/jacs.7b07603] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The histone deacetylase family comprises 18 enzymes that catalyze deacetylation of acetylated lysine residues; however, the specificity and substrate profile of each isozyme remains largely unknown. Due to transient enzyme-substrate interactions, conventional co-immunoprecipitation methods frequently fail to identify enzyme-specific substrates. Additionally, compensatory mechanisms often limit the ability of knockdown or chemical inhibition studies to achieve significant fold changes observed by acetylation proteomics methods. Furthermore, measured alterations do not guarantee a direct link between enzyme and substrate. Here we present a chemical crosslinking strategy that incorporates a photoreactive, non-natural amino acid, p-benzoyl-l-phenylalanine, into various positions of the structurally characterized isozyme histone deacetylase 8 (HDAC8). After covalent capture, co-immunoprecipitation, and mass spectrometric analysis, we identified a subset of HDAC8 substrates from human cell lysates, which were further validated for catalytic turnover. Overall, this chemical crosslinking approach identified novel HDAC8-specific substrates with high catalytic efficiency, thus presenting a general strategy for unbiased deacetylase substrate discovery.
Collapse
Affiliation(s)
- Jeffrey E Lopez
- Program in Chemical Biology, ‡Department of Chemistry, and §Department of Biological Chemistry, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Sarah E Haynes
- Program in Chemical Biology, ‡Department of Chemistry, and §Department of Biological Chemistry, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Jaimeen D Majmudar
- Program in Chemical Biology, ‡Department of Chemistry, and §Department of Biological Chemistry, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Brent R Martin
- Program in Chemical Biology, ‡Department of Chemistry, and §Department of Biological Chemistry, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Carol A Fierke
- Program in Chemical Biology, ‡Department of Chemistry, and §Department of Biological Chemistry, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
30
|
HDAC8 functions in spindle assembly during mouse oocyte meiosis. Oncotarget 2017; 8:20092-20102. [PMID: 28223544 PMCID: PMC5386746 DOI: 10.18632/oncotarget.15383] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/17/2017] [Indexed: 12/19/2022] Open
Abstract
HDAC8 is a class I histone deacetylase that functions in a variety of biological processes through its non-histone substrates. However, its roles during oocyte meiosis remain elusive. Here, we document that HDAC8 localizes at spindle poles and positively participates in the regulation of microtubule organization and spindle assembly in mouse oocytes. Depletion of HDAC8 by siRNA-based gene silencing results in various spindle defects and chromosome misalignment during oocyte meiotic maturation, accompanied by impaired kinetochore-microtubule attachments. Consequently, a higher incidence of aneuploidy is generated in HDAC8-depleted MII eggs. In addition, inhibition of HDAC8 activity with its selective inhibitor PCI-34051 phenocopies the spindle/chromosome defects resulting from HDAC8 depletion by siRNA injection. Finally, we find that HDAC8 is required for the correct localization of ϕ-tubulin to spindle poles. Collectively, these data reveal that HDAC8 plays a significant role in regulating spindle assembly and thus ensuring the euploidy in mouse eggs.
Collapse
|
31
|
Castaneda CA, Lopez JE, Joseph CG, Scholle MD, Mrksich M, Fierke CA. Active Site Metal Identity Alters Histone Deacetylase 8 Substrate Selectivity: A Potential Novel Regulatory Mechanism. Biochemistry 2017; 56:5663-5670. [PMID: 28937750 DOI: 10.1021/acs.biochem.7b00851] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Histone deacetylase 8 (HDAC8) is a well-characterized member of the class I acetyl-lysine deacetylase (HDAC) family. Previous work has shown that the efficiency of HDAC8-catalyzed deacetylation of a methylcoumarin peptide varies depending on the identity of the divalent metal ion in the HDAC8 active site. Here we demonstrate that both HDAC8 activity and substrate selectivity for a diverse range of peptide substrates depend on the identity of the active site metal ion. Varied deacetylase activities of Fe(II)- and Zn(II)-HDAC8 toward an array of peptide substrates were identified using self-assembled monolayers for matrix-assisted laser desorption ionization (SAMDI) mass spectrometry. Subsequently, the metal dependence of deacetylation of peptides of biological interest was measured using an in vitro peptide assay. While Fe(II)-HDAC8 is generally more active than Zn(II)-HDAC8, the Fe(II)/Zn(II) HDAC8 activity ratio varies widely (from 2 to 150) among the peptides tested. These data provide support for the hypothesis that HDAC8 may undergo metal switching in vivo that, in turn, may regulate its activity. However, future studies are needed to explore the identity of the metal ion bound to HDAC8 in cells under varied conditions.
Collapse
Affiliation(s)
- Carol Ann Castaneda
- Program in Chemical Biology, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Jeffrey E Lopez
- Program in Chemical Biology, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Caleb G Joseph
- Department of Medicinal Chemistry, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Michael D Scholle
- Department of Chemistry and Department of Biomedical Engineering, Northwestern University , Evanston, Illinois 60208, United States
| | - Milan Mrksich
- Department of Chemistry and Department of Biomedical Engineering, Northwestern University , Evanston, Illinois 60208, United States
| | - Carol A Fierke
- Program in Chemical Biology, University of Michigan , Ann Arbor, Michigan 48109, United States.,Department of Medicinal Chemistry, University of Michigan , Ann Arbor, Michigan 48109, United States.,Department of Chemistry and Department of Biological Chemistry, University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
32
|
Evolutionary relationships among protein lysine deacetylases of parasites causing neglected diseases. INFECTION GENETICS AND EVOLUTION 2017; 53:175-188. [DOI: 10.1016/j.meegid.2017.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 05/10/2017] [Accepted: 05/12/2017] [Indexed: 12/20/2022]
|
33
|
Marcu O, Dodson EJ, Alam N, Sperber M, Kozakov D, Lensink MF, Schueler-Furman O. FlexPepDock lessons from CAPRI peptide-protein rounds and suggested new criteria for assessment of model quality and utility. Proteins 2017; 85:445-462. [PMID: 28002624 PMCID: PMC6618814 DOI: 10.1002/prot.25230] [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: 09/13/2016] [Revised: 11/15/2016] [Accepted: 11/23/2016] [Indexed: 12/21/2022]
Abstract
CAPRI rounds 28 and 29 included, for the first time, peptide-receptor targets of three different systems, reflecting increased appreciation of the importance of peptide-protein interactions. The CAPRI rounds allowed us to objectively assess the performance of Rosetta FlexPepDock, one of the first protocols to explicitly include peptide flexibility in docking, accounting for peptide conformational changes upon binding. We discuss here successes and challenges in modeling these targets: we obtain top-performing, high-resolution models of the peptide motif for cases with known binding sites but there is a need for better modeling of flanking regions, as well as better selection criteria, in particular for unknown binding sites. These rounds have also provided us the opportunity to reassess the success criteria, to better reflect the quality of a peptide-protein complex model. Using all models submitted to CAPRI, we analyze the correlation between current classification criteria and the ability to retrieve critical interface features, such as hydrogen bonds and hotspots. We find that loosening the backbone (and ligand) RMSD threshold, together with a restriction on the side chain RMSD measure, allows us to improve the selection of high-accuracy models. We also suggest a new measure to assess interface hydrogen bond recovery, which is not assessed by the current CAPRI criteria. Finally, we find that surprisingly much can be learned from rather inaccurate models about binding hotspots, suggesting that the current status of peptide-protein docking methods, as reflected by the submitted CAPRI models, can already have a significant impact on our understanding of protein interactions. Proteins 2017; 85:445-462. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Orly Marcu
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| | - Emma-Joy Dodson
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| | - Nawsad Alam
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| | - Michal Sperber
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brooks University, Stony Brook, New York, 11794
| | - Marc F Lensink
- University of Lille, CNRS UMR8576 UGSF, Lille, 59000, France
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| |
Collapse
|
34
|
Mukhtar YM, Huang Y, Liu J, Chen D, Zheng W. Acetanilide and bromoacetyl-lysine derivatives as activators for human histone deacetylase 8. Bioorg Med Chem Lett 2017; 27:2319-2323. [DOI: 10.1016/j.bmcl.2017.04.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/08/2017] [Accepted: 04/12/2017] [Indexed: 11/27/2022]
|
35
|
Modeling Peptide-Protein Structure and Binding Using Monte Carlo Sampling Approaches: Rosetta FlexPepDock and FlexPepBind. Methods Mol Biol 2017; 1561:139-169. [PMID: 28236237 DOI: 10.1007/978-1-4939-6798-8_9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Many signaling and regulatory processes involve peptide-mediated protein interactions, i.e., the binding of a short stretch in one protein to a domain in its partner. Computational tools that generate accurate models of peptide-receptor structures and binding improve characterization and manipulation of known interactions, help to discover yet unknown peptide-protein interactions and networks, and bring into reach the design of peptide-based drugs for targeting specific systems of medical interest.Here, we present a concise overview of the Rosetta FlexPepDock protocol and its derivatives that we have developed for the structure-based characterization of peptide-protein binding. Rosetta FlexPepDock was built to generate precise models of protein-peptide complex structures, by effectively addressing the challenge of the considerable conformational flexibility of the peptide. Rosetta FlexPepBind is an extension of this protocol that allows characterizing peptide-binding affinities and specificities of various biological systems, based on the structural models generated by Rosetta FlexPepDock. We provide detailed descriptions and guidelines for the usage of these protocols, and on a specific example, we highlight the variety of different challenges that can be met and the questions that can be answered with Rosetta FlexPepDock.
Collapse
|
36
|
Targeting histone deacetylase 8 as a therapeutic approach to cancer and neurodegenerative diseases. Future Med Chem 2016; 8:1609-34. [PMID: 27572818 DOI: 10.4155/fmc-2016-0117] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Histone deacetylase 8 (HDAC8), a unique class I zinc-dependent HDAC, is an emerging target in cancer and other diseases. Its substrate repertoire extends beyond histones to many nonhistone proteins. Besides being a deacetylase, HDAC8 also mediates signaling via scaffolding functions. Aberrant expression or deregulated interactions with transcription factors are critical in HDAC8-dependent cancers. Many potent HDAC8-selective inhibitors with cellular activity and anticancer effects have been reported. We present HDAC8 as a druggable target and discuss inhibitors of different chemical scaffolds with cellular effects. Furthermore, we review HDAC8 activators that revert activity of mutant enzymes. Isotype-selective HDAC8 targeting in patients with HDAC8-relevant cancers is challenging, however, is promising to avoid adverse side effects as observed with pan-HDAC inhibitors.
Collapse
|