1
|
Oliveira-Silva JM, Oliveira LS, Chiminazo CB, Fonseca R, de Souza CVE, Aissa AF, de Almeida Lima GD, Ionta M, Castro-Gamero AM. WT161, a selective HDAC6 inhibitor, decreases growth, enhances chemosensitivity, promotes apoptosis, and suppresses motility of melanoma cells. Cancer Chemother Pharmacol 2025; 95:22. [PMID: 39821335 DOI: 10.1007/s00280-024-04731-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: 09/01/2024] [Accepted: 11/25/2024] [Indexed: 01/19/2025]
Abstract
PURPOSE Histone deacetylase 6 (HDAC6) plays a critical role in tumorigenesis and tumor progression, contributing to proliferation, chemoresistance, and cell motility by regulating microtubule architecture. Despite its upregulation in melanoma tissues and cell lines, the specific biological roles of HDAC6 in melanoma are not well understood. This study aims to explore the functional effects and underlying mechanisms of WT161, a selective HDAC6 inhibitor, in melanoma cell lines. METHODS Cell proliferation was assessed using both 2D and 3D cell culture systems, including MTT assays, spheroid growth analyses, and colony formation assays. The interaction between WT161 and the chemotherapeutic agents temozolomide (TMZ) or dacarbazine (DTIC) was evaluated using the Chou-Talalay method. Apoptotic cell death was analyzed through flow cytometry, while migration, adhesion, and invasion assays were conducted to evaluate the motility capacities of melanoma cells. Western blot assays quantified α-tubulin acetylation (Lys40), PARP cleavage, and protein levels of β-catenin and E-cadherin. RESULTS WT161 significantly reduced cell growth in both 2D and 3D cultures, decreased clonogenic capacity, and showed synergistic interactions with TMZ and DTIC. The inhibitor also induced apoptotic cell death and enhanced TMZ-induced apoptosis. Additionally, WT161 reduced cell migration and invasion while increasing cell adhesion. These effects were linked to changes in β-catenin and E-cadherin levels, depending on the specific cell type evaluated. CONCLUSION Our study underscores the pivotal role of HDAC6 in melanoma progression, establishing it as a promising therapeutic target. We provide the first comprehensive evidence of WT161's anti-melanoma effects, setting the stage for further research into HDAC6 inhibitors as a potential strategy for melanoma treatment.
Collapse
Affiliation(s)
- João Marcos Oliveira-Silva
- Human Genetics Laboratory, Institute of Natural Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
- Postgraduate Program in Biosciences Applied to Health (PPGB), Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
| | - Leilane Sales Oliveira
- Human Genetics Laboratory, Institute of Natural Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
- Postgraduate Program in Biosciences Applied to Health (PPGB), Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
| | - Carolina Berraut Chiminazo
- Human Genetics Laboratory, Institute of Natural Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
| | - Rafael Fonseca
- Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
| | | | - Alexandre Ferro Aissa
- Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
- Postgraduate Program in Biosciences Applied to Health (PPGB), Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
| | - Graziela Domingues de Almeida Lima
- Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
- Postgraduate Program in Biosciences Applied to Health (PPGB), Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
| | - Marisa Ionta
- Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
- Postgraduate Program in Biosciences Applied to Health (PPGB), Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil
| | - Angel Mauricio Castro-Gamero
- Human Genetics Laboratory, Institute of Natural Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil.
- Postgraduate Program in Biosciences Applied to Health (PPGB), Federal University of Alfenas (UNIFAL-MG), Alfenas, MG, 37130-001, Brazil.
| |
Collapse
|
2
|
Andreu-Carbó M, Egoldt C, Aumeier C. Microtubule shaft integrity emerges as a crucial determinant of the acetylation pattern. Cytoskeleton (Hoboken) 2025; 82:55-57. [PMID: 38923402 DOI: 10.1002/cm.21887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Affiliation(s)
- Mireia Andreu-Carbó
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Cornelia Egoldt
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Charlotte Aumeier
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| |
Collapse
|
3
|
Kravec M, Šedo O, Nedvědová J, Micka M, Šulcová M, Zezula N, Gömöryová K, Potěšil D, Sri Ganji R, Bologna S, Červenka I, Zdráhal Z, Harnoš J, Tripsianes K, Janke C, Bařinka C, Bryja V. Carboxy-terminal polyglutamylation regulates signaling and phase separation of the Dishevelled protein. EMBO J 2024; 43:5635-5666. [PMID: 39349846 PMCID: PMC11574253 DOI: 10.1038/s44318-024-00254-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/15/2024] [Accepted: 09/16/2024] [Indexed: 11/20/2024] Open
Abstract
Polyglutamylation is a reversible posttranslational modification that is catalyzed by enzymes of the tubulin tyrosine ligase-like (TTLL) family. Here, we found that TTLL11 generates a previously unknown type of polyglutamylation that is initiated by the addition of a glutamate residue to the free C-terminal carboxyl group of a substrate protein. TTLL11 efficiently polyglutamylates the Wnt signaling protein Dishevelled 3 (DVL3), thereby changing the interactome of DVL3. Polyglutamylation increases the capacity of DVL3 to get phosphorylated, to undergo phase separation, and to act in the noncanonical Wnt pathway. Both carboxy-terminal polyglutamylation and the resulting reduction in phase separation capacity of DVL3 can be reverted by the deglutamylating enzyme CCP6, demonstrating a causal relationship between TTLL11-mediated polyglutamylation and phase separation. Thus, C-terminal polyglutamylation represents a new type of posttranslational modification, broadening the range of proteins that can be modified by polyglutamylation and providing the first evidence that polyglutamylation can modulate protein phase separation.
Collapse
Affiliation(s)
- Marek Kravec
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Ondrej Šedo
- Central European Institute of Technology (CEITEC), Brno, Czech Republic
| | - Jana Nedvědová
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Miroslav Micka
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Central European Institute of Technology (CEITEC), Brno, Czech Republic
| | - Marie Šulcová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Nikodém Zezula
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Kristína Gömöryová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - David Potěšil
- Central European Institute of Technology (CEITEC), Brno, Czech Republic
| | - Ranjani Sri Ganji
- Central European Institute of Technology (CEITEC), Brno, Czech Republic
| | - Sara Bologna
- Central European Institute of Technology (CEITEC), Brno, Czech Republic
| | - Igor Červenka
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute of Technology (CEITEC), Brno, Czech Republic
| | - Jakub Harnoš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | - Carsten Janke
- Institut Curie, Université PSL, CNRS UMR3348, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Cyril Bařinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
4
|
Sun S, Xu Z, He L, Shen Y, Yan Y, Lv X, Zhu X, Li W, Tian WY, Zheng Y, Lin S, Sun Y, Li L. Metabolic regulation of cytoskeleton functions by HDAC6-catalyzed α-tubulin lactylation. Nat Commun 2024; 15:8377. [PMID: 39333081 PMCID: PMC11437170 DOI: 10.1038/s41467-024-52729-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Posttranslational modifications (PTMs) of tubulin, termed the "tubulin code", play important roles in regulating microtubule functions within subcellular compartments for specialized cellular activities. While numerous tubulin PTMs have been identified, a comprehensive understanding of the complete repertoire is still underway. In this study, we report that α-tubulin lactylation is catalyzed by HDAC6 by using lactate to increase microtubule dynamics in neurons. We identify lactylation on lysine 40 of α-tubulin in the soluble tubulin dimers. Notably, lactylated α-tubulin enhances microtubule dynamics and facilitates neurite outgrowth and branching in cultured hippocampal neurons. Moreover, we discover an unexpected function of HDAC6, acting as the primary lactyltransferase to catalyze α-tubulin lactylation. HDAC6-catalyzed lactylation is a reversible process, dependent on lactate concentrations. Intracellular lactate concentration triggers HDAC6 to lactylate α-tubulin, a process dependent on its deacetylase activity. Additionally, the lactyltransferase activity may be conserved in HDAC family proteins. Our study reveals the primary role of HDAC6 in regulating α-tubulin lactylation, establishing a link between cell metabolism and cytoskeleton functions.
Collapse
Affiliation(s)
- Shuangshuang Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhe Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Liying He
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yihui Shen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuqing Yan
- Department of Pain management, HuaDong Hospital Affiliated to Fudan University, Shanghai, China
| | - Xubing Lv
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xujing Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wei Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, China
| | - Wei-Ya Tian
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yongjun Zheng
- Department of Pain management, HuaDong Hospital Affiliated to Fudan University, Shanghai, China
| | - Sen Lin
- Department of Neurology, 2nd Affiliated Hospital, Army Medical University, Chongqing, China
| | - Yadong Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lei Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
5
|
van Eyll J, Prior R, Celanire S, Van Den Bosch L, Rombouts F. Therapeutic indications for HDAC6 inhibitors in the peripheral and central nervous disorders. Expert Opin Ther Targets 2024; 28:719-737. [PMID: 39305025 DOI: 10.1080/14728222.2024.2404571] [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: 04/16/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Inhibition of the enzymatic function of HDAC6 is currently being explored in clinical trials ranging from peripheral neuropathies to cancers. Advances in selective HDAC6 inhibitor discovery allowed studying highly efficacious brain penetrant and peripheral restrictive compounds for treating PNS and CNS indications. AREAS COVERED This review explores the multifactorial role of HDAC6 in cells, the common pathological hallmarks of PNS and CNS disorders, and how HDAC6 modulates these mechanisms. Pharmacological inhibition of HDAC6 and genetic knockout/knockdown studies as a therapeutic strategy in PNS and CNS indications were analyzed. Furthermore, we describe the recent developments in HDAC6 PET tracers and their utility in CNS indications. Finally, we explore the advancements and challenges with HDAC6 inhibitor compounds, such as hydroxamic acid, fluoromethyl oxadiazoles, HDAC6 degraders, and thiol-based inhibitors. EXPERT OPINION Based on extensive preclinical evidence, pharmacological inhibition of HDAC6 is a promising approach for treating both PNS and CNS disorders, given its involvement in neurodegeneration and aging-related cellular processes. Despite the progress in the development of selective HDAC6 inhibitors, safety concerns remain regarding their chronic administration in PNS and CNS indications, and the development of novel compound classes and modalities inhibiting HDAC6 function offer a way to mitigate some of these safety concerns.
Collapse
Affiliation(s)
| | | | - Sylvain Celanire
- Augustine Therapeutics, Research and Development, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | | |
Collapse
|
6
|
Chen J, Zehr EA, Gruschus JM, Szyk A, Liu Y, Tanner ME, Tjandra N, Roll-Mecak A. Tubulin code eraser CCP5 binds branch glutamates by substrate deformation. Nature 2024; 631:905-912. [PMID: 39020174 DOI: 10.1038/s41586-024-07699-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 06/11/2024] [Indexed: 07/19/2024]
Abstract
Microtubule function is modulated by the tubulin code, diverse posttranslational modifications that are altered dynamically by writer and eraser enzymes1. Glutamylation-the addition of branched (isopeptide-linked) glutamate chains-is the most evolutionarily widespread tubulin modification2. It is introduced by tubulin tyrosine ligase-like enzymes and erased by carboxypeptidases of the cytosolic carboxypeptidase (CCP) family1. Glutamylation homeostasis, achieved through the balance of writers and erasers, is critical for normal cell function3-9, and mutations in CCPs lead to human disease10-13. Here we report cryo-electron microscopy structures of the glutamylation eraser CCP5 in complex with the microtubule, and X-ray structures in complex with transition-state analogues. Combined with NMR analysis, these analyses show that CCP5 deforms the tubulin main chain into a unique turn that enables lock-and-key recognition of the branch glutamate in a cationic pocket that is unique to CCP family proteins. CCP5 binding of the sequences flanking the branch point primarily through peptide backbone atoms enables processing of diverse tubulin isotypes and non-tubulin substrates. Unexpectedly, CCP5 exhibits inefficient processing of an abundant β-tubulin isotype in the brain. This work provides an atomistic view into glutamate branch recognition and resolution, and sheds light on homeostasis of the tubulin glutamylation syntax.
Collapse
Affiliation(s)
- Jiayi Chen
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Elena A Zehr
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - James M Gruschus
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Agnieszka Szyk
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Yanjie Liu
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin E Tanner
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nico Tjandra
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
7
|
Mahale A, Routholla G, Lavanya S, Sharma P, Ghosh B, Kulkarni OP. Pharmacological blockade of HDAC6 attenuates cancer progression by inhibiting IL-1β and modulating immunosuppressive response in OSCC. Int Immunopharmacol 2024; 132:111921. [PMID: 38547770 DOI: 10.1016/j.intimp.2024.111921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 05/01/2024]
Abstract
Interleukin-1-beta (IL-1β) one of the biomarkers for oral squamous cell carcinoma (OSCC), is upregulated in tumor-microenvironment (TME) and associated with poor patient survival. Thus, a novel modulator of IL-1β would be of great therapeutic value for OSCC treatment. Here we report regulation of IL-1β and TME by histone deacetylase-6 (HDAC6)-inhibitor in OSCC. We observed significant upregulation of HDAC6 in 4-nitroquniline (4-NQO)-induced OSCC in mice and 4-NQO & Lipopolysaccharide (LPS) stimulated OSCC and fibroblast cells. Tubastatin A (TSA)-attenuated the OSCC progression in mice as observed improvement in the histology over tongue and esophagus, with reduced tumor burden. TSA treatment to 4-NQO mice attenuated protein expression of HDAC6, pro-and-mature-IL-1β and pro-and-cleaved-caspase-1 and ameliorated acetylated-tubulin. In support of our experimental work, human TCGA analysis revealed HDAC6 and IL-1β were upregulated in the primary tumor, with different tumor stages and grades. We found TSA modulate TME, indicated by downregulation of CD11b+Gr1+-Myeloid-derived suppressor cells, CD11b+F4/80+CD206+ M2-macrophages and increase in CD11b+F4/80+MHCII+ M1-macrophages. TSA significantly reduced the gene expression of HDAC6, IL-1β, Arginase-1 and iNOS in isolated splenic-MDSCs. FaDu-HTB-43 and NIH3T3 cells stimulated with LPS and 4-NQO exhibit higher IL-1β levels in the supernatant. Interestingly, immunoblot analysis of the cell lysate, we observed that TSA does not alter the expression as well as activation of IL-1β and caspase-1 but the acetylated-tubulin was found to be increased. Nocodazole pre-treatment proved that TSA inhibited the lysosomal exocytosis of IL-1β through tubulin acetylation. In conclusion, HDAC6 inhibitors attenuated TME and cancer progression through the regulation of IL-1β in OSCC.
Collapse
Affiliation(s)
- Ashutosh Mahale
- Metabolic Disorders and Neuroscience Research laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Ganesh Routholla
- Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - S Lavanya
- Metabolic Disorders and Neuroscience Research laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Pravesh Sharma
- Metabolic Disorders and Neuroscience Research laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Onkar Prakash Kulkarni
- Metabolic Disorders and Neuroscience Research laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India.
| |
Collapse
|
8
|
Andreu-Carbó M, Egoldt C, Velluz MC, Aumeier C. Microtubule damage shapes the acetylation gradient. Nat Commun 2024; 15:2029. [PMID: 38448418 PMCID: PMC10918088 DOI: 10.1038/s41467-024-46379-5] [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: 01/09/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
The properties of single microtubules within the microtubule network can be modulated through post-translational modifications (PTMs), including acetylation within the lumen of microtubules. To access the lumen, the enzymes could enter through the microtubule ends and at damage sites along the microtubule shaft. Here we show that the acetylation profile depends on damage sites, which can be caused by the motor protein kinesin-1. Indeed, the entry of the deacetylase HDAC6 into the microtubule lumen can be modulated by kinesin-1-induced damage sites. In contrast, activity of the microtubule acetylase αTAT1 is independent of kinesin-1-caused shaft damage. On a cellular level, our results show that microtubule acetylation distributes in an exponential gradient. This gradient results from tight regulation of microtubule (de)acetylation and scales with the size of the cells. The control of shaft damage represents a mechanism to regulate PTMs inside the microtubule by giving access to the lumen.
Collapse
Affiliation(s)
| | - Cornelia Egoldt
- Department of Biochemistry, University of Geneva, 1211, Geneva, Switzerland
| | | | - Charlotte Aumeier
- Department of Biochemistry, University of Geneva, 1211, Geneva, Switzerland.
| |
Collapse
|
9
|
Chu JC, Tseng HJ, Lee SB, Hsu KC, Hsin LW, Liang RH, Lin TE, Gao NC, Chen LC, Lu WH, Wang AHJ, Huang WJ. Synthesis and biological evaluation of C-4 substituted phenoxazine-bearing hydroxamic acids with potent class II histone deacetylase inhibitory activities. J Enzyme Inhib Med Chem 2023; 38:2212326. [PMID: 37190931 DOI: 10.1080/14756366.2023.2212326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
Class II histone deacetylases (HDACs) are considered as potential targets to treat Alzheimer's disease (AD). Previously, C-3 substituted phenothiazine-containing compounds with class II HDAC-inhibiting activities was found to promote neurite outgrowth. This study replaced phenothiazine moiety with phenoxazine that contains many C-3 and C-4 substituents. Some resulting compounds bearing the C-4 substituent on a phenoxazine ring displayed potent class II HDAC inhibitory activities. Structure-activity relationship (SAR) of these compounds that inhibited HDAC isoenzymes was disclosed. Molecular modelling analysis demonstrates that the potent activities of C-4 substituted compounds probably arise from π-π stacked interactions between these compounds and class IIa HDAC enzymes. One of these, compound 7d exhibited the most potent class II HDAC inhibition (IC50= 3-870 nM). Notably, it protected neuron cells from H2O2-induced neuron damage at sub-μM concentrations, but with no significant cytotoxicity. These findings show that compound 7d is a lead compound for further development of anti-neurodegenerative agents.
Collapse
Affiliation(s)
- Jung-Chun Chu
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Hui-Ju Tseng
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, United States
| | - Sung-Bau Lee
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kai-Cheng Hsu
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Ling-Wei Hsin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ru-Hao Liang
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Nain-Chu Gao
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Liang-Chieh Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California, United States
| | - Wan-Hsun Lu
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Andrew H-J Wang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Jan Huang
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
10
|
McKenna ED, Sarbanes SL, Cummings SW, Roll-Mecak A. The Tubulin Code, from Molecules to Health and Disease. Annu Rev Cell Dev Biol 2023; 39:331-361. [PMID: 37843925 DOI: 10.1146/annurev-cellbio-030123-032748] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Microtubules are essential dynamic polymers composed of α/β-tubulin heterodimers. They support intracellular trafficking, cell division, cellular motility, and other essential cellular processes. In many species, both α-tubulin and β-tubulin are encoded by multiple genes with distinct expression profiles and functionality. Microtubules are further diversified through abundant posttranslational modifications, which are added and removed by a suite of enzymes to form complex, stereotyped cellular arrays. The genetic and chemical diversity of tubulin constitute a tubulin code that regulates intrinsic microtubule properties and is read by cellular effectors, such as molecular motors and microtubule-associated proteins, to provide spatial and temporal specificity to microtubules in cells. In this review, we synthesize the rapidly expanding tubulin code literature and highlight limitations and opportunities for the field. As complex microtubule arrays underlie essential physiological processes, a better understanding of how cells employ the tubulin code has important implications for human disease ranging from cancer to neurological disorders.
Collapse
Affiliation(s)
- Elizabeth D McKenna
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Stephanie L Sarbanes
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Steven W Cummings
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA;
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
11
|
Calogero AM, Basellini MJ, Isilgan HB, Longhena F, Bellucci A, Mazzetti S, Rolando C, Pezzoli G, Cappelletti G. Acetylated α-Tubulin and α-Synuclein: Physiological Interplay and Contribution to α-Synuclein Oligomerization. Int J Mol Sci 2023; 24:12287. [PMID: 37569662 PMCID: PMC10418364 DOI: 10.3390/ijms241512287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Emerging evidence supports that altered α-tubulin acetylation occurs in Parkinson's disease (PD), a neurodegenerative disorder characterized by the deposition of α-synuclein fibrillary aggregates within Lewy bodies and nigrostriatal neuron degeneration. Nevertheless, studies addressing the interplay between α-tubulin acetylation and α-synuclein are lacking. Here, we investigated the relationship between α-synuclein and microtubules in primary midbrain murine neurons and the substantia nigra of post-mortem human brains. Taking advantage of immunofluorescence and Proximity Ligation Assay (PLA), a method allowing us to visualize protein-protein interactions in situ, combined with confocal and super-resolution microscopy, we found that α-synuclein and acetylated α-tubulin colocalized and were in close proximity. Next, we employed an α-synuclein overexpressing cellular model and tested the role of α-tubulin acetylation in α-synuclein oligomer formation. We used the α-tubulin deacetylase HDAC6 inhibitor Tubacin to modulate α-tubulin acetylation, and we evaluated the presence of α-synuclein oligomers by PLA. We found that the increase in acetylated α-tubulin significantly induced α-synuclein oligomerization. In conclusion, we unraveled the link between acetylated α-tubulin and α-synuclein and demonstrated that α-tubulin acetylation could trigger the early step of α-synuclein aggregation. These data suggest that the proper regulation of α-tubulin acetylation might be considered a therapeutic strategy to take on PD.
Collapse
Affiliation(s)
- Alessandra Maria Calogero
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
- Fondazione Grigioni per il Morbo di Parkinson, 20125 Milan, Italy;
| | - Milo Jarno Basellini
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
| | - Huseyin Berkcan Isilgan
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
| | - Francesca Longhena
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (F.L.); (A.B.)
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (F.L.); (A.B.)
| | - Samanta Mazzetti
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
- Fondazione Grigioni per il Morbo di Parkinson, 20125 Milan, Italy;
| | - Chiara Rolando
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
| | - Gianni Pezzoli
- Fondazione Grigioni per il Morbo di Parkinson, 20125 Milan, Italy;
- Parkinson Institute, ASST-Pini-CTO, 20126 Milan, Italy
| | - Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (M.J.B.); (H.B.I.); (S.M.); (C.R.)
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
12
|
Cellupica E, Caprini G, Fossati G, Mirdita D, Cordella P, Marchini M, Rocchio I, Sandrone G, Stevenazzi A, Vergani B, Steinkühler C, Vanoni MA. The Importance of the "Time Factor" for the Evaluation of Inhibition Mechanisms: The Case of Selected HDAC6 Inhibitors. BIOLOGY 2023; 12:1049. [PMID: 37626935 PMCID: PMC10452033 DOI: 10.3390/biology12081049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023]
Abstract
Histone deacetylases (HDACs) participate with histone acetyltransferases in the modulation of the biological activity of a broad array of proteins, besides histones. Histone deacetylase 6 is unique among HDAC as it contains two catalytic domains, an N-terminal microtubule binding region and a C-terminal ubiquitin binding domain. Most of its known biological roles are related to its protein lysine deacetylase activity in the cytoplasm. The design of specific inhibitors is the focus of a large number of medicinal chemistry programs in the academy and industry because lowering HDAC6 activity has been demonstrated to be beneficial for the treatment of several diseases, including cancer, and neurological and immunological disorders. Here, we show how re-evaluation of the mechanism of action of selected HDAC6 inhibitors, by monitoring the time-dependence of the onset and relief of the inhibition, revealed instances of slow-binding/slow-release inhibition. The same approach, in conjunction with X-ray crystallography, in silico modeling and mass spectrometry, helped to propose a model of inhibition of HDAC6 by a novel difluoromethyloxadiazole-based compound that was found to be a slow-binding substrate analog of HDAC6, giving rise to a tightly bound, long-lived inhibitory derivative.
Collapse
Affiliation(s)
- Edoardo Cellupica
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | - Gianluca Caprini
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | - Gianluca Fossati
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | - Doris Mirdita
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milano, Italy;
| | - Paola Cordella
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | - Mattia Marchini
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | - Ilaria Rocchio
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | - Giovanni Sandrone
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | - Andrea Stevenazzi
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | - Barbara Vergani
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | - Christian Steinkühler
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092 Cinisello Balsamo, Italy; (E.C.); (G.C.); (G.F.); (P.C.); (M.M.); (I.R.); (G.S.); (A.S.); (B.V.)
| | | |
Collapse
|
13
|
Motlová L, Šnajdr I, Kutil Z, Andris E, Ptáček J, Novotná A, Nováková Z, Havlínová B, Tueckmantel W, Dráberová H, Majer P, Schutkowski M, Kozikowski A, Rulíšek L, Bařinka C. Comprehensive Mechanistic View of the Hydrolysis of Oxadiazole-Based Inhibitors by Histone Deacetylase 6 (HDAC6). ACS Chem Biol 2023. [PMID: 37392419 PMCID: PMC10367051 DOI: 10.1021/acschembio.3c00212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2023]
Abstract
Histone deacetylase (HDAC) inhibitors used in the clinic typically contain a hydroxamate zinc-binding group (ZBG). However, more recent work has shown that the use of alternative ZBGs, and, in particular, the heterocyclic oxadiazoles, can confer higher isoenzyme selectivity and more favorable ADMET profiles. Herein, we report on the synthesis and biochemical, crystallographic, and computational characterization of a series of oxadiazole-based inhibitors selectively targeting the HDAC6 isoform. Surprisingly, but in line with a very recent finding reported in the literature, a crystal structure of the HDAC6/inhibitor complex revealed that hydrolysis of the oxadiazole ring transforms the parent oxadiazole into an acylhydrazide through a sequence of two hydrolytic steps. An identical cleavage pattern was also observed both in vitro using the purified HDAC6 enzyme as well as in cellular systems. By employing advanced quantum and molecular mechanics (QM/MM) and QM calculations, we elucidated the mechanistic details of the two hydrolytic steps to obtain a comprehensive mechanistic view of the double hydrolysis of the oxadiazole ring. This was achieved by fully characterizing the reaction coordinate, including identification of the structures of all intermediates and transition states, together with calculations of their respective activation (free) energies. In addition, we ruled out several (intuitively) competing pathways. The computed data (ΔG‡ ≈ 21 kcal·mol-1 for the rate-determining step of the overall dual hydrolysis) are in very good agreement with the experimentally determined rate constants, which a posteriori supports the proposed reaction mechanism. We also clearly (and quantitatively) explain the role of the -CF3 or -CHF2 substituent on the oxadiazole ring, which is a prerequisite for hydrolysis to occur. Overall, our data provide compelling evidence that the oxadiazole warheads can be efficiently transformed within the active sites of target metallohydrolases to afford reaction products possessing distinct selectivity and inhibition profiles.
Collapse
Affiliation(s)
- Lucia Motlová
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Ivan Šnajdr
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10 Prague 6, Czech Republic
| | - Zsófia Kutil
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Erik Andris
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10 Prague 6, Czech Republic
| | - Jakub Ptáček
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Adéla Novotná
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10 Prague 6, Czech Republic
| | - Zora Nováková
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Barbora Havlínová
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Werner Tueckmantel
- StarWise Therapeutics LLC, University Research Park, Inc., Madison, Wisconsin 53719, United States
| | - Helena Dráberová
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10 Prague 6, Czech Republic
| | - Mike Schutkowski
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | - Alan Kozikowski
- StarWise Therapeutics LLC, University Research Park, Inc., Madison, Wisconsin 53719, United States
| | - Lubomír Rulíšek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10 Prague 6, Czech Republic
| | - Cyril Bařinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| |
Collapse
|
14
|
Chen J, Roll-Mecak A. Glutamylation is a negative regulator of microtubule growth. Mol Biol Cell 2023; 34:ar70. [PMID: 37074962 PMCID: PMC10295482 DOI: 10.1091/mbc.e23-01-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/20/2023] Open
Abstract
Microtubules are noncovalent polymers built from αβ-tubulin dimers. The disordered C-terminal tubulin tails are functionalized with multiple glutamate chains of variable lengths added and removed by tubulin tyrosine ligases (TTLLs) and carboxypeptidases (CCPs). Glutamylation is abundant on stable microtubule arrays such as in axonemes and axons, and its dysregulation leads to human pathologies. Despite this, the effects of glutamylation on intrinsic microtubule dynamics are unclear. Here we generate tubulin with short and long glutamate chains and show that glutamylation slows the rate of microtubule growth and increases catastrophes as a function of glutamylation levels. This implies that the higher stability of glutamylated microtubules in cells is due to effectors. Interestingly, EB1 is minimally affected by glutamylation and thus can report on the growth rates of both unmodified and glutamylated microtubules. Finally, we show that glutamate removal by CCP1 and 5 is synergistic and occurs preferentially on soluble tubulin, unlike TTLL enzymes that prefer microtubules. This substrate preference establishes an asymmetry whereby once the microtubule depolymerizes, the released tubulin is reset to a less-modified state, while polymerized tubulin accumulates the glutamylation mark. Our work shows that a modification on the disordered tubulin tails can directly affect microtubule dynamics and furthers our understanding of the mechanistic underpinnings of the tubulin code.
Collapse
Affiliation(s)
- Jiayi Chen
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, and
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, and
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD 20892
| |
Collapse
|
15
|
Iwanski MK, Kapitein LC. Cellular cartography: Towards an atlas of the neuronal microtubule cytoskeleton. Front Cell Dev Biol 2023; 11:1052245. [PMID: 37035244 PMCID: PMC10073685 DOI: 10.3389/fcell.2023.1052245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Microtubules, one of the major components of the cytoskeleton, play a crucial role during many aspects of neuronal development and function, such as neuronal polarization and axon outgrowth. Consequently, the microtubule cytoskeleton has been implicated in many neurodevelopmental and neurodegenerative disorders. The polar nature of microtubules is quintessential for their function, allowing them to serve as tracks for long-distance, directed intracellular transport by kinesin and dynein motors. Most of these motors move exclusively towards either the plus- or minus-end of a microtubule and some have been shown to have a preference for either dynamic or stable microtubules, those bearing a particular post-translational modification or those decorated by a specific microtubule-associated protein. Thus, it becomes important to consider the interplay of these features and their combinatorial effects on transport, as well as how different types of microtubules are organized in the cell. Here, we discuss microtubule subsets in terms of tubulin isotypes, tubulin post-translational modifications, microtubule-associated proteins, microtubule stability or dynamicity, and microtubule orientation. We highlight techniques used to study these features of the microtubule cytoskeleton and, using the information from these studies, try to define the composition, role, and organization of some of these subsets in neurons.
Collapse
Affiliation(s)
| | - Lukas C. Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
16
|
Ptacek J, Snajdr I, Schimer J, Kutil Z, Mikesova J, Baranova P, Havlinova B, Tueckmantel W, Majer P, Kozikowski A, Barinka C. Selectivity of Hydroxamate- and Difluoromethyloxadiazole-Based Inhibitors of Histone Deacetylase 6 In Vitro and in Cells. Int J Mol Sci 2023; 24:4720. [PMID: 36902164 PMCID: PMC10003107 DOI: 10.3390/ijms24054720] [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: 02/06/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023] Open
Abstract
Histone deacetylase 6 (HDAC6) is a unique member of the HDAC family of enzymes due to its complex domain organization and cytosolic localization. Experimental data point toward the therapeutic use of HDAC6-selective inhibitors (HDAC6is) for use in both neurological and psychiatric disorders. In this article, we provide side-by-side comparisons of hydroxamate-based HDAC6is frequently used in the field and a novel HDAC6 inhibitor containing the difluoromethyl-1,3,4-oxadiazole function as an alternative zinc-binding group (compound 7). In vitro isotype selectivity screening uncovered HDAC10 as a primary off-target for the hydroxamate-based HDAC6is, while compound 7 features exquisite 10,000-fold selectivity over all other HDAC isoforms. Complementary cell-based assays using tubulin acetylation as a surrogate readout revealed approximately 100-fold lower apparent potency for all compounds. Finally, the limited selectivity of a number of these HDAC6is is shown to be linked to cytotoxicity in RPMI-8226 cells. Our results clearly show that off-target effects of HDAC6is must be considered before attributing observed physiological readouts solely to HDAC6 inhibition. Moreover, given their unparalleled specificity, the oxadiazole-based inhibitors would best be employed either as research tools in further probing HDAC6 biology or as leads in the development of truly HDAC6-specific compounds in the treatment of human disease states.
Collapse
Affiliation(s)
- Jakub Ptacek
- Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Ivan Snajdr
- Institute of Organic Chemistry and Biochemistry of the Academy of Sciences of the Czech Republic, Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Jiri Schimer
- Institute of Organic Chemistry and Biochemistry of the Academy of Sciences of the Czech Republic, Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Zsofia Kutil
- Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Jana Mikesova
- Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Petra Baranova
- Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Barbora Havlinova
- Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Werner Tueckmantel
- StarWise Therapeutics LLC, University Research Park, Inc., Madison, WI 53719, USA
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry of the Academy of Sciences of the Czech Republic, Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Alan Kozikowski
- StarWise Therapeutics LLC, University Research Park, Inc., Madison, WI 53719, USA
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Cyril Barinka
- Institute of Biotechnology CAS, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| |
Collapse
|
17
|
Shukla S, Komarek J, Novakova Z, Nedvedova J, Ustinova K, Vankova P, Kadek A, Uetrecht C, Mertens H, Barinka C. In-solution structure and oligomerization of human histone deacetylase 6 - an integrative approach. FEBS J 2023; 290:821-836. [PMID: 36062318 DOI: 10.1111/febs.16616] [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: 11/05/2021] [Revised: 07/08/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023]
Abstract
Human histone deacetylase 6 (HDAC6) is a structurally unique, multidomain protein implicated in a variety of physiological processes including cytoskeletal remodelling and the maintenance of cellular homeostasis. Our current understanding of the HDAC6 structure is limited to isolated domains, and a holistic picture of the full-length protein structure, including possible domain interactions, is missing. Here, we used an integrative structural biology approach to build a solution model of HDAC6 by combining experimental data from several orthogonal biophysical techniques complemented by molecular modelling. We show that HDAC6 is best described as a mosaic of folded and intrinsically disordered domains that in-solution adopts an ensemble of conformations without any stable interactions between structured domains. Furthermore, HDAC6 forms dimers/higher oligomers in a concentration-dependent manner, and its oligomerization is mediated via the positively charged N-terminal microtubule-binding domain. Our findings provide the first insights into the structure of full-length human HDAC6 and can be used as a basis for further research into structure function and physiological studies of this unique deacetylase.
Collapse
Affiliation(s)
- Shivam Shukla
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic.,Department of Physical Chemistry, Faculty of Natural Science, Charles University, Prague, Czech Republic
| | - Jan Komarek
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Zora Novakova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Jana Nedvedova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Kseniya Ustinova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Pavla Vankova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Alan Kadek
- Leibniz Institute of Virology (LIV), Hamburg, Germany.,European XFEL GmbH, Schenefeld, Germany
| | - Charlotte Uetrecht
- Leibniz Institute of Virology (LIV), Hamburg, Germany.,European XFEL GmbH, Schenefeld, Germany.,Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron (DESY), Hamburg, Germany.,Department of Health Sciences and Biomedicine, School of Life Sciences, University of Siegen, Germany
| | - Haydyn Mertens
- European Molecular Biology Laboratory (EMBL)-Hamburg Outstation, c/o DESY, Germany
| | - Cyril Barinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
18
|
Wattanathamsan O, Chantaravisoot N, Wongkongkathep P, Kungsukool S, Chetprayoon P, Chanvorachote P, Vinayanuwattikun C, Pongrakhananon V. Inhibition of histone deacetylase 6 destabilizes ERK phosphorylation and suppresses cancer proliferation via modulation of the tubulin acetylation-GRP78 interaction. J Biomed Sci 2023; 30:4. [PMID: 36639650 PMCID: PMC9838051 DOI: 10.1186/s12929-023-00898-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The leading cause of cancer-related mortality worldwide is lung cancer, and its clinical outcome and prognosis are still unsatisfactory. The understanding of potential molecular targets is necessary for clinical implications in precision diagnostic and/or therapeutic purposes. Histone deacetylase 6 (HDAC6), a major deacetylase enzyme, is a promising target for cancer therapy; however, the molecular mechanism regulating cancer pathogenesis is largely unknown. METHODS The clinical relevance of HDAC6 expression levels and their correlation with the overall survival rate were analyzed based on the TCGA and GEO databases. HDAC6 expression in clinical samples obtained from lung cancer tissues and patient-derived primary lung cancer cells was evaluated using qRT-PCR and Western blot analysis. The potential regulatory mechanism of HDAC6 was identified by proteomic analysis and validated by immunoblotting, immunofluorescence, microtubule sedimentation, and immunoprecipitation-mass spectrometry (IP-MS) assays using a specific inhibitor of HDAC6, trichostatin A (TSA) and RNA interference to HDAC6 (siHDAC6). Lung cancer cell growth was assessed by an in vitro 2-dimensional (2D) cell proliferation assay and 3D tumor spheroid formation using patient-derived lung cancer cells. RESULTS HDAC6 was upregulated in lung cancer specimens and significantly correlated with poor prognosis. Inhibition of HDAC6 by TSA and siHDAC6 caused downregulation of phosphorylated extracellular signal-regulated kinase (p-ERK), which was dependent on the tubulin acetylation status. Tubulin acetylation induced by TSA and siHDAC6 mediated the dissociation of p-ERK on microtubules, causing p-ERK destabilization. The proteomic analysis demonstrated that the molecular chaperone glucose-regulated protein 78 (GRP78) was an important scaffolder required for p-ERK localization on microtubules, and this phenomenon was significantly inhibited by either TSA, siHDAC6, or siGRP78. In addition, suppression of HDAC6 strongly attenuated an in vitro 2D lung cancer cell growth and an in vitro 3D patient derived-lung cancer spheroid growth. CONCLUSIONS HDAC6 inhibition led to upregulate tubulin acetylation, causing GRP78-p-ERK dissociation from microtubules. As a result, p-ERK levels were decreased, and lung cancer cell growth was subsequently suppressed. This study reveals the intriguing role and molecular mechanism of HDAC6 as a tumor promoter, and its inhibition represents a promising approach for anticancer therapy.
Collapse
Affiliation(s)
- Onsurang Wattanathamsan
- grid.7922.e0000 0001 0244 7875Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences,, Chulalongkorn University, Bangkok, Thailand
| | - Naphat Chantaravisoot
- grid.7922.e0000 0001 0244 7875Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand ,grid.7922.e0000 0001 0244 7875Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Piriya Wongkongkathep
- grid.7922.e0000 0001 0244 7875Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sakkarin Kungsukool
- grid.413637.40000 0004 4682 905XDepartment of Respiratory Medicine, Central Chest Institute of Thailand, Muang District, Nonthaburi, Thailand
| | - Paninee Chetprayoon
- grid.425537.20000 0001 2191 4408Toxicology and Bio Evaluation Service Center, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Pithi Chanvorachote
- grid.7922.e0000 0001 0244 7875Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences,, Chulalongkorn University, Bangkok, Thailand
| | - Chanida Vinayanuwattikun
- grid.7922.e0000 0001 0244 7875Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Varisa Pongrakhananon
- grid.7922.e0000 0001 0244 7875Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences,, Chulalongkorn University, Bangkok, Thailand ,grid.7922.e0000 0001 0244 7875Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Cluster, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
19
|
Mikesova J, Ondrakova M, Jelinkova I, Ptacek J, Novakova Z, Barinka C. Determining Potency of Inhibitors Targeting Histone Deacetylase 6 by Quantification of Acetylated Tubulin in Cells. Methods Mol Biol 2023; 2589:455-466. [PMID: 36255642 DOI: 10.1007/978-1-0716-2788-4_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
During the preclinical development of small molecule inhibitors, compounds or compound libraries are typically first screened using purified target enzymes in vitro to select candidates with high potency. In the later stages of the development, however, functional cell-based assays may provide biologically more relevant data. In this chapter, we describe a detailed protocol for determining the potency of inhibitors targeting human histone deacetylase 6 in complex cellular environments. Cells are first treated with a dilution series of tested compounds, cell lysates separated by SDS-PAGE, and electrotransferred to a blotting membrane. The inhibitor potency is then determined indirectly by quantifying the levels of acetylated tubulin as a surrogate readout.
Collapse
Affiliation(s)
- Jana Mikesova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Marketa Ondrakova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Iva Jelinkova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Jakub Ptacek
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Zora Novakova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Cyril Barinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic.
| |
Collapse
|
20
|
Abstract
Heart disease remains the leading cause of morbidity and mortality worldwide. With the advancement of modern technology, the role(s) of microtubules in the pathogenesis of heart disease has become increasingly apparent, though currently there are limited treatments targeting microtubule-relevant mechanisms. Here, we review the functions of microtubules in the cardiovascular system and their specific adaptive and pathological phenotypes in cardiac disorders. We further explore the use of microtubule-targeting drugs and highlight promising druggable therapeutic targets for the future treatment of heart diseases.
Collapse
Affiliation(s)
- Emily F Warner
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, United Kingdom (E.F.W., X.L.)
| | - Yang Li
- Department of Cardiovascular Surgery, Zhongnan Hospital, Wuhan University School of Medicine, People's Republic of China (Y.L.)
| | - Xuan Li
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, United Kingdom (E.F.W., X.L.)
| |
Collapse
|
21
|
Kutil Z, Meleshin M, Baranova P, Havlinova B, Schutkowski M, Barinka C. Characterization of the class IIa histone deacetylases substrate specificity. FASEB J 2022; 36:e22287. [PMID: 35349187 DOI: 10.1096/fj.202101663r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/07/2022] [Accepted: 03/17/2022] [Indexed: 11/11/2022]
Abstract
Class IIa histone deacetylases (HDACs) play critical roles in vertebrate development and physiology, yet direct evidence of their intrinsic deacetylase activity and on substrate specificity regarding the peptide sequence is still missing. In this study, we designed and synthesized a combinatorial peptide library allowing us to profile class IIa HDACs sequence specificity at positions +3 through -3 from the central lysine modified by the well-accepted trifluoroacetyl function. Our data revealed a strong preference for bulky aromatic acids directly flanking the central trifluoroacetyllysine, while all class IIa HDACs disfavor positively charged residues and proline at the +1/-1 positions. The chemical nature of amino acid residues N-terminally to the central trifluoroacetyllysine has a more profound effect on substrate recognition as compared to residues located C-terminally. These findings were validated by designing selected favored and disfavored peptide sequences, with the favored ones are accepted with catalytic efficacy of 75 000 and 525 000 M-1 s-1 for HDAC7 and HDAC5, respectively. Results reported here could help in developing class IIa HDACs inhibitors and also in the search for new natural class IIa HDACs substrates.
Collapse
Affiliation(s)
- Zsofia Kutil
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Marat Meleshin
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Petra Baranova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Barbora Havlinova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Mike Schutkowski
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Cyril Barinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
22
|
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
|
23
|
Yuan W, Liu B, Sanda M, Wei R, Benicky J, Novakova Z, Barinka C, Goldman R. Glycoforms of human prostate-specific membrane antigen (PSMA) in human cells and prostate tissue. Prostate 2022; 82:132-144. [PMID: 34662441 PMCID: PMC9646948 DOI: 10.1002/pros.24254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/27/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION N-glycosylation is a ubiquitous and variable posttranslational modification that regulates physiological functions of secretory and membrane-associated proteins and the dysregulation of glycosylation pathways is often associated with cancer growth and metastasis. Prostate-specific membrane antigen (PSMA) is an established biomarker for prostate cancer imaging and therapy. METHODS Mass spectrometry was used to analyze the distribution of the site-specific glycoforms of PSMA in insect, human embryonic kidney, and prostate cancer cells, and in prostate tissue upon immunoaffinity enrichment. RESULTS While recombinant PSMA expressed in insect cells was decorated mainly by paucimannose and high mannose glycans, complex, hybrid, and high mannose glycans were detected in samples from human cells and tissue. We noted an interesting spatial distribution of the glycoforms on the PSMA surface-high mannose glycans were the dominant glycoforms at the N459, N476, and N638 sequons facing the plasma membrane, while the N121, N195, and N336 sites, located at the exposed apical PSMA domain, carried primarily complex glycans. The presence of high mannose glycoforms at the former sequons likely results from the limited access of enzymes of the glycosynthetic pathway required for the synthesis of the complex structures. In line with the limited accessibility of membrane-proximal sites, no glycosylation was observed at the N51 site positioned closest to the membrane. CONCLUSIONS Our study presents initial descriptive analysis of the glycoforms of PSMA observed in cell lines and in prostate tissue. It will hopefully stimulate further research into PSMA glycoforms in the context of tumor staging, noninvasive detection of prostate tumors, and the impact of glycoforms on physicochemical and enzymatic characteristics of PSMA in a tissue-specific manner.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
- Clinical and Translational Glycoscience Research Center, Georgetown University Medical Center, Georgetown University, Washington DC, USA
| | - Baoqin Liu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| | - Miloslav Sanda
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
- Clinical and Translational Glycoscience Research Center, Georgetown University Medical Center, Georgetown University, Washington DC, USA
| | - Renhuizi Wei
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
- Clinical and Translational Glycoscience Research Center, Georgetown University Medical Center, Georgetown University, Washington DC, USA
| | - Julius Benicky
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
- Clinical and Translational Glycoscience Research Center, Georgetown University Medical Center, Georgetown University, Washington DC, USA
| | - Zora Novakova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Cyril Barinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Radoslav Goldman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
- Clinical and Translational Glycoscience Research Center, Georgetown University Medical Center, Georgetown University, Washington DC, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington DC, USA
| |
Collapse
|
24
|
Novakova Z, Khuntsaria D, Gresova M, Mikesova J, Havlinova B, Shukla S, Kolarova L, Vesela K, Martasek P, Barinka C. Heterologous expression and purification of recombinant human protoporphyrinogen oxidase IX: A comparative study. PLoS One 2021; 16:e0259837. [PMID: 34793488 PMCID: PMC8601502 DOI: 10.1371/journal.pone.0259837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022] Open
Abstract
Human protoporphyrinogen oxidase IX (hPPO) is an oxygen-dependent enzyme catalyzing the penultimate step in the heme biosynthesis pathway. Mutations in the enzyme are linked to variegate porphyria, an autosomal dominant metabolic disease. Here we investigated eukaryotic cells as alternative systems for heterologous expression of hPPO, as the use of a traditional bacterial-based system failed to produce several clinically relevant hPPO variants. Using bacterially-produced hPPO, we first analyzed the impact of N-terminal tags and various detergent on hPPO yield, and specific activity. Next, the established protocol was used to compare hPPO constructs heterologously expressed in mammalian HEK293T17 and insect Hi5 cells with prokaryotic overexpression. By attaching various fusion partners at the N- and C-termini of hPPO we also evaluated the influence of the size and positioning of fusion partners on expression levels, specific activity, and intracellular targeting of hPPO fusions in mammalian cells. Overall, our results suggest that while enzymatically active hPPO can be heterologously produced in eukaryotic systems, the limited availability of the intracellular FAD co-factor likely negatively influences yields of a correctly folded protein making thus the E.coli a system of choice for recombinant hPPO overproduction. At the same time, PPO overexpression in eukaryotic cells might be preferrable in cases when the effects of post-translational modifications (absent in bacteria) on target protein functions are studied.
Collapse
Affiliation(s)
- Zora Novakova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Daria Khuntsaria
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Marketa Gresova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Jana Mikesova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Barbora Havlinova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Shivam Shukla
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Lucie Kolarova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Katerina Vesela
- First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Pavel Martasek
- First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Cyril Barinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
- * E-mail:
| |
Collapse
|
25
|
In vivo evaluation of a microtubule PET ligand, [ 11C]MPC-6827, in mice following chronic alcohol consumption. Pharmacol Rep 2021; 74:241-247. [PMID: 34491568 DOI: 10.1007/s43440-021-00311-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Excessive alcohol consumption is a global health burden and requires a better understanding of its neurobiology. A lower density of brain microtubules is found in alcohol-related human brain disease postmortem and in rodent models of chronic alcohol consumption. Here, we report in vivo imaging studies of microtubules in brain using our recently reported Positron Emission Tomography (PET) tracer, [11C]MPC-6827, in chronic alcohol-consuming adult male C57BL/6 J mice and control mice. METHODS In vivo PET imaging studies of [11C]MPC-6827 (3.7 ± 0.8 MBq) were performed in two groups of adult male mice: (1) water-consuming control mice (n = 4) and (2) mice that consumed 20% alcohol (w/v) for 4 months using the intermittent 2-bottle choice procedure that has been shown to lead to signs of alcohol dependence. Dynamic 63 min PET images were acquired using a microPET Inveon system (Siemens, Germany). PET images were reconstructed using the 3D-OSEM algorithm and analyzed using VivoQuant version 4 (Invicro, MA). Tracer uptake in ROIs that included whole brain, prefrontal cortex (PFC), liver and heart was measured and plotted as %ID/g over time (0-63 min) to generate time-activity curves (TACs). RESULTS In general, a trend for lower binding of [11C]MPC-6827 in the whole brain and PFC of mice in the chronic alcohol group was found compared with control group. No group difference in radiotracer binding was found in the peripheral organs such as liver and heart. CONCLUSIONS This pilot study indicates a trend of loss of microtubule binding in whole brain and prefrontal cortex of chronic alcohol administered mice brain compared to control mice, but no loss in heart or liver. These results indicate the potential of [11C]MPC-6827 as a PET ligand for further in vivo imaging investigations of AUD in human.
Collapse
|
26
|
English K, Barton MC. HDAC6: A Key Link Between Mitochondria and Development of Peripheral Neuropathy. Front Mol Neurosci 2021; 14:684714. [PMID: 34531721 PMCID: PMC8438325 DOI: 10.3389/fnmol.2021.684714] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/30/2021] [Indexed: 01/21/2023] Open
Abstract
Peripheral neuropathy, which is the result of nerve damage from lesions or disease, continues to be a major health concern due to the common manifestation of neuropathic pain. Most investigations into the development of peripheral neuropathy focus on key players such as voltage-gated ion channels or glutamate receptors. However, emerging evidence points to mitochondrial dysfunction as a major player in the development of peripheral neuropathy and resulting neuropathic pain. Mitochondrial dysfunction in neuropathy includes altered mitochondrial transport, mitochondrial metabolism, as well as mitochondrial dynamics. The mechanisms that lead to mitochondrial dysfunction in peripheral neuropathy are poorly understood, however, the Class IIb histone deacetylase (HDAC6), may play an important role in the process. HDAC6 is a key regulator in multiple mechanisms of mitochondrial dynamics and may contribute to mitochondrial dysregulation in peripheral neuropathy. Accumulating evidence shows that HDAC6 inhibition is strongly associated with alleviating peripheral neuropathy and neuropathic pain, as well as mitochondrial dysfunction, in in vivo and in vitro models of peripheral neuropathy. Thus, HDAC6 inhibitors are being investigated as potential therapies for multiple peripheral neuropathic disorders. Here, we review emerging studies and integrate recent advances in understanding the unique connection between peripheral neuropathy and mitochondrial dysfunction through HDAC6-mediated interactions.
Collapse
Affiliation(s)
- Krystal English
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- UTHealth McGovern Medical School, Houston, TX, United States
| | - Michelle Craig Barton
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
27
|
Nardella F, Halby L, Dobrescu I, Viluma J, Bon C, Claes A, Cadet-Daniel V, Tafit A, Roesch C, Hammam E, Erdmann D, Mairet-Khedim M, Peronet R, Mecheri S, Witkowski B, Scherf A, Arimondo PB. Procainamide-SAHA Fused Inhibitors of hHDAC6 Tackle Multidrug-Resistant Malaria Parasites. J Med Chem 2021; 64:10403-10417. [PMID: 34185525 DOI: 10.1021/acs.jmedchem.1c00821] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epigenetic post-translational modifications are essential for human malaria parasite survival and progression through its life cycle. Here, we present new functionalized suberoylanilide hydroxamic acid (SAHA) derivatives that chemically combine the pan-histone deacetylase inhibitor SAHA with the DNA methyltransferase inhibitor procainamide. A three- or four-step chemical synthesis was designed starting from cheap raw materials. Compared to the single drugs, the combined molecules showed a superior activity in Plasmodium and a potent inhibition against human HDAC6, exerting no cytotoxicity in human cell lines. These new compounds are fully active in multidrug-resistant Plasmodium falciparum Cambodian isolates. They target transmission of the parasite by inducing irreversible morphological changes in gametocytes and inhibiting exflagellation. The compounds are slow-acting and have an additive antimalarial effect in combination with fast-acting epidrugs and dihydroartemisinin. The lead compound decreases parasitemia in mice in a severe malaria model. Taken together, this novel fused molecule offers an affordable alternative to current failing antimalarial therapy.
Collapse
Affiliation(s)
- Flore Nardella
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Ludovic Halby
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| | - Irina Dobrescu
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Johanna Viluma
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| | - Corentin Bon
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France.,Ecole Doctorale MTCI ED563, Université de Paris, Sorbonne Paris Cité, Paris 75270, France
| | - Aurélie Claes
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Véronique Cadet-Daniel
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| | - Ambre Tafit
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| | - Camille Roesch
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Elie Hammam
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Diane Erdmann
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France.,Ecole Doctorale MTCI ED563, Université de Paris, Sorbonne Paris Cité, Paris 75270, France
| | - Melissa Mairet-Khedim
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Roger Peronet
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Salah Mecheri
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Benoit Witkowski
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Artur Scherf
- Unité Biologie des Interactions Hôte-Parasite, Département de Parasites et Insectes Vecteurs, Institut Pasteur, CNRS ERL 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Paola B Arimondo
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, UMR n°3523, CNRS, 28 Rue du Dr Roux, Paris 75015, France
| |
Collapse
|
28
|
Doyle TM, Salvemini D. Mini-Review: Mitochondrial dysfunction and chemotherapy-induced neuropathic pain. Neurosci Lett 2021; 760:136087. [PMID: 34182057 DOI: 10.1016/j.neulet.2021.136087] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a somatosensory axonopathy in cancer patients receiving any of a variety of widely-use antitumor agents. CIPN can lead to long-lasting neuropathic pain that limits the dose or length of otherwise life-saving cancer therapy. Accumulating evidence over the last two decades indicates that many chemotherapeutic agents cause mitochondrial injury in the peripheral sensory nerves by disrupting mitochondrial structure and bioenergetics, increasing nitro-oxidative stress and altering mitochondrial transport, fission, fusion and mitophagy. The accumulation of abnormal and dysfunctional mitochondria in sensory neurons are linked to axonal growth defects resulting in the loss of intraepidermal nerve fibers in the hands and feet, increased spontaneous discharge and the sensitization of peripheral sensory neurons that provoke and promote changes in the central nervous system that establish a chronic neuropathic pain state. This has led to the propose mitotoxicity theory of CIPN. Strategies that improve mitochondrial function have shown success in preventing and reversing CIPN in pre-clinical animal models and have begun to show some progress toward translation to the clinic. In this review, we will review the evidence for, the causes and effects of and current strategies to target mitochondrial dysfunction in CIPN.
Collapse
Affiliation(s)
- Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO 63104, USA; Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO 63104, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO 63104, USA; Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO 63104, USA.
| |
Collapse
|
29
|
Skwarska A, Calder EDD, Sneddon D, Bolland H, Odyniec ML, Mistry IN, Martin J, Folkes LK, Conway SJ, Hammond EM. Development and pre-clinical testing of a novel hypoxia-activated KDAC inhibitor. Cell Chem Biol 2021; 28:1258-1270.e13. [PMID: 33910023 PMCID: PMC8460716 DOI: 10.1016/j.chembiol.2021.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/15/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
Tumor hypoxia is associated with therapy resistance and poor patient prognosis. Hypoxia-activated prodrugs, designed to selectively target hypoxic cells while sparing normal tissue, represent a promising treatment strategy. We report the pre-clinical efficacy of 1-methyl-2-nitroimidazole panobinostat (NI-Pano, CH-03), a novel bioreductive version of the clinically used lysine deacetylase inhibitor, panobinostat. NI-Pano was stable in normoxic (21% O2) conditions and underwent NADPH-CYP-mediated enzymatic bioreduction to release panobinostat in hypoxia (<0.1% O2). Treatment of cells grown in both 2D and 3D with NI-Pano increased acetylation of histone H3 at lysine 9, induced apoptosis, and decreased clonogenic survival. Importantly, NI-Pano exhibited growth delay effects as a single agent in tumor xenografts. Pharmacokinetic analysis confirmed the presence of sub-micromolar concentrations of panobinostat in hypoxic mouse xenografts, but not in circulating plasma or kidneys. Together, our pre-clinical results provide a strong mechanistic rationale for the clinical development of NI-Pano for selective targeting of hypoxic tumors.
Collapse
Affiliation(s)
- Anna Skwarska
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Ewen D D Calder
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Deborah Sneddon
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Hannah Bolland
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Maria L Odyniec
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Ishna N Mistry
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Jennifer Martin
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Lisa K Folkes
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Stuart J Conway
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK.
| | - Ester M Hammond
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| |
Collapse
|
30
|
Cappelletti G, Calogero AM, Rolando C. Microtubule acetylation: A reading key to neural physiology and degeneration. Neurosci Lett 2021; 755:135900. [PMID: 33878428 DOI: 10.1016/j.neulet.2021.135900] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 02/02/2023]
Abstract
Neurons are the perfect example of cells where microtubules are essential to achieve an extraordinary degree of morphological and functional complexity. Different tubulin isoforms and associated post-translational modifications are the basis to establish the diversity in biochemical and biophysical properties of microtubules including their stability and the control of intracellular transport. Acetylation is one of the key tubulin modifications and it can influence important structural, mechanical and biological traits of the microtubule network. Here, we present the emerging evidence for the essential role of microtubule acetylation in the control of neuronal and glial function in healthy and degenerative conditions. In particular, we discuss the pathogenic role of tubulin acetylation in neurodegenerative disorders and focus on Parkinson's disease. We also provide a critical analysis about the possibility to target tubulin acetylation as a novel therapeutic intervention for neuroprotective strategies.
Collapse
Affiliation(s)
- Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, Milano, Italy; Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milano, Italy.
| | | | - Chiara Rolando
- Department of Biosciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
31
|
Shen S, Picci C, Ustinova K, Benoy V, Kutil Z, Zhang G, Tavares MT, Pavlíček J, Zimprich CA, Robers MB, Van Den Bosch L, Bařinka C, Langley B, Kozikowski AP. Tetrahydroquinoline-Capped Histone Deacetylase 6 Inhibitor SW-101 Ameliorates Pathological Phenotypes in a Charcot-Marie-Tooth Type 2A Mouse Model. J Med Chem 2021; 64:4810-4840. [PMID: 33830764 DOI: 10.1021/acs.jmedchem.0c02210] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Histone deacetylase 6 (HDAC6) is a promising therapeutic target for the treatment of neurodegenerative disorders. SW-100 (1a), a phenylhydroxamate-based HDAC6 inhibitor (HDAC6i) bearing a tetrahydroquinoline (THQ) capping group, is a highly potent and selective HDAC6i that was shown to be effective in mouse models of Fragile X syndrome and Charcot-Marie-Tooth disease type 2A (CMT2A). In this study, we report the discovery of a new THQ-capped HDAC6i, termed SW-101 (1s), that possesses excellent HDAC6 potency and selectivity, together with markedly improved metabolic stability and druglike properties compared to SW-100 (1a). X-ray crystallography data reveal the molecular basis of HDAC6 inhibition by SW-101 (1s). Importantly, we demonstrate that SW-101 (1s) treatment elevates the impaired level of acetylated α-tubulin in the distal sciatic nerve, counteracts progressive motor dysfunction, and ameliorates neuropathic symptoms in a CMT2A mouse model bearing mutant MFN2. Taken together, these results bode well for the further development of SW-101 (1s) as a disease-modifying HDAC6i.
Collapse
Affiliation(s)
- Sida Shen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Cristina Picci
- School of Health, The University of Waikato, Private Bag 3105, Hamilton 3240, New Zealand
| | - Kseniya Ustinova
- Institute of Biotechnology of the Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | - Veronick Benoy
- Laboratory of Neurobiology, Center for Brain & Disease (VIB) and Leuven Brain Institute (LBI), KU Leuven, B-3000 Leuven, Belgium
| | - Zsófia Kutil
- Institute of Biotechnology of the Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | - Guiping Zhang
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Maurício T Tavares
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Jiří Pavlíček
- Institute of Biotechnology of the Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | - Chad A Zimprich
- Promega Corporation, Madison, Wisconsin 53711, United States
| | | | - Ludo Van Den Bosch
- Laboratory of Neurobiology, Center for Brain & Disease (VIB) and Leuven Brain Institute (LBI), KU Leuven, B-3000 Leuven, Belgium
| | - Cyril Bařinka
- Institute of Biotechnology of the Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | - Brett Langley
- School of Health, The University of Waikato, Private Bag 3105, Hamilton 3240, New Zealand
| | | |
Collapse
|
32
|
Hsu KC, Chu JC, Tseng HJ, Liu CI, Wang HC, Lin TE, Lee HS, Hsin LW, Wang AHJ, Lin CH, Huang WJ. Synthesis and biological evaluation of phenothiazine derivative-containing hydroxamic acids as potent class II histone deacetylase inhibitors. Eur J Med Chem 2021; 219:113419. [PMID: 33845233 DOI: 10.1016/j.ejmech.2021.113419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/22/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022]
Abstract
The pathogenesis of Alzheimer's disease (AD) has been associated with dysregulation of histone deacetylases (HDACs). Previously, acridine-based HDAC inhibitors have shown potential in ameliorating HDAC activity and enhancing neurite outgrowth. In this study, the acridine ring was modified using various phenothiazine derivatives. Several resulting compounds exhibited potent enzyme-inhibiting activity towards class II HDACs when compared to the clinically approved HDAC inhibitor SAHA. Compound 4f demonstrated the highest class II HDAC inhibition (IC50 = 4.6-600 nM), as well as promotion of neurite outgrowth. Importantly, compound 4f displayed no cytotoxicity against neuron cells. Compound 4f was further evaluated for cellular effects. Altogether, these findings show a potential strategy in HDAC inhibition for treatment of the neurological disease.
Collapse
Affiliation(s)
- Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taiwan; Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jung-Chun Chu
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Hui-Ju Tseng
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chia-I Liu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hao-Ching Wang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Master Program in Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hong-Sheng Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ling-Wei Hsin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Andrew H-J Wang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chien-Huang Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Wei-Jan Huang
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
33
|
Yang WT, Hong SR, He K, Ling K, Shaiv K, Hu J, Lin YC. The Emerging Roles of Axonemal Glutamylation in Regulation of Cilia Architecture and Functions. Front Cell Dev Biol 2021; 9:622302. [PMID: 33748109 PMCID: PMC7970040 DOI: 10.3389/fcell.2021.622302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cilia, which either generate coordinated motion or sense environmental cues and transmit corresponding signals to the cell body, are highly conserved hair-like structures that protrude from the cell surface among diverse species. Disruption of ciliary functions leads to numerous human disorders, collectively referred to as ciliopathies. Cilia are mechanically supported by axonemes, which are composed of microtubule doublets. It has been recognized for several decades that tubulins in axonemes undergo glutamylation, a post-translational polymodification, that conjugates glutamic acid chains onto the C-terminal tail of tubulins. However, the physiological roles of axonemal glutamylation were not uncovered until recently. This review will focus on how cells modulate glutamylation on ciliary axonemes and how axonemal glutamylation regulates cilia architecture and functions, as well as its physiological importance in human health. We will also discuss the conventional and emerging new strategies used to manipulate glutamylation in cilia.
Collapse
Affiliation(s)
- Wen-Ting Yang
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
| | - Shi-Rong Hong
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
| | - Kai He
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Kritika Shaiv
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
| | - JingHua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, HsinChu City, Taiwan
- Department of Medical Science, National Tsing Hua University, HsinChu City, Taiwan
| |
Collapse
|
34
|
Prejanò M, Vidossich P, Russo N, De Vivo M, Marino T. Insights into the Catalytic Mechanism of Domains CD1 and CD2 in Histone Deacetylase 6 from Quantum Calculations. ACS Catal 2021. [DOI: 10.1021/acscatal.0c04729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Mario Prejanò
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Via Ponte Pietro Bucci, 87036 Arcavacata di Rende, Cosenza, Italy
| | - Pietro Vidossich
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Nino Russo
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Via Ponte Pietro Bucci, 87036 Arcavacata di Rende, Cosenza, Italy
| | - Marco De Vivo
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Tiziana Marino
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Via Ponte Pietro Bucci, 87036 Arcavacata di Rende, Cosenza, Italy
| |
Collapse
|
35
|
Ružić D, Đoković N, Nikolić K, Vujić Z. Medicinal chemistry of histone deacetylase inhibitors. ARHIV ZA FARMACIJU 2021. [DOI: 10.5937/arhfarm71-30618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Today, we are witnessing an explosion of scientific concepts in cancer chemotherapy. It has been considered for a long time that genetic instability in cancer should be treated with drugs that directly damage the DNA. Understanding the molecular basis of malignant diseases shed light on studying phenotypic plasticity. In the era of epigenetics, many efforts are being made to alter the aberrant homeostasis in cancer without modifying the DNA sequence. One such strategy is modulation of the lysine acetylome in human cancers. To remove the acetyl group from the histones, cells use the enzymes that are called histone deacetylases (HDACs). The disturbed equilibrium between acetylation and deacetylation on lysine residues of histones can be manipulated with histone deacetylase inhibitors (HDACi). Throughout the review, an effort will be made to present the mechanistic basis of targeting the HDAC isoforms, discovered selective HDAC inhibitors, and their therapeutical implications and expectations in modern drug discovery.
Collapse
|
36
|
Ha N, Choi YI, Jung N, Song JY, Bae DK, Kim MC, Lee YJ, Song H, Kwak G, Jeong S, Park S, Nam SH, Jung S, Choi B. A novel histone deacetylase 6 inhibitor improves myelination of Schwann cells in a model of Charcot-Marie-Tooth disease type 1A. Br J Pharmacol 2020; 177:5096-5113. [PMID: 33460073 PMCID: PMC7589015 DOI: 10.1111/bph.15231] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 07/22/2020] [Accepted: 07/25/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Charcot-Marie-Tooth (CMT) disease is the most common hereditary peripheral neuropathy. CMT type 1A (CMT1A) accounts for approximately 50% of CMT patients and is linked to PMP22 gene duplication. Histone deacetylase-6 (HDAC6) has pleiotropic effects, such as regulating lipid homeostasis and cellular stress. Although HDAC6 has been regarded as a promising drug target for neurodegenerative diseases, its inhibition has not yet been tested in CMT1A. Here we have tested the therapeutic potential of CKD-504, a clinical stage HDAC6 inhibitor, in a mouse model of CMT1A EXPERIMENTAL APPROACH: The potency and selectivity of CKD-504 was evaluated, using a HDAC enzyme panel assay and western blots. The therapeutic potential of CKD-504 was evaluated using behavioural testing and electrophysiological assessments in the C22 mouse model of CMT1A. PMP22 protein expression and aggregation were analysed in mesenchymal stem cell-derived Schwann cells from CMT1A patients and sciatic nerves from C22 mice. KEY RESULTS The HDAC6 inhibitor, CKD-504, modulated molecular chaperon proteins such as HSP90 and HSP70, which are involved in the folding/refolding of proteins such as PMP22. CKD-504 treatment restored myelination in both mesenchymal stem cell-derived Schwann cells from CMT1A patients and sciatic nerves of C22 mice and improved the axonal integrity of the sciatic nerve, leading to behavioural, electrophysiological, and histological improvements in C22 mice. CONCLUSION AND IMPLICATIONS A novel HDAC6 inhibitor, CKD-504, has potent therapeutic efficacy for CMT1A.
Collapse
Affiliation(s)
- Nina Ha
- Department of Health Sciences and Technology, SAIHSTSungkyunkwan UniversitySeoulRepublic of Korea
- CKD Research InstituteYonginRepublic of Korea
| | | | - Namhee Jung
- Department of Biochemistry, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| | | | | | | | | | | | - Geon Kwak
- Department of Health Sciences and Technology, SAIHSTSungkyunkwan UniversitySeoulRepublic of Korea
| | - Soyeon Jeong
- Department of Biochemistry, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| | - Saeyoung Park
- Department of Biochemistry, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| | - Soo Hyun Nam
- Department of Neurology, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Sung‐Chul Jung
- Department of Biochemistry, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| | - Byung‐Ok Choi
- Department of Health Sciences and Technology, SAIHSTSungkyunkwan UniversitySeoulRepublic of Korea
- Department of Neurology, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
| |
Collapse
|
37
|
Microtubule Dysfunction: A Common Feature of Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21197354. [PMID: 33027950 PMCID: PMC7582320 DOI: 10.3390/ijms21197354] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/24/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons are particularly susceptible to microtubule (MT) defects and deregulation of the MT cytoskeleton is considered to be a common insult during the pathogenesis of neurodegenerative disorders. Evidence that dysfunctions in the MT system have a direct role in neurodegeneration comes from findings that several forms of neurodegenerative diseases are associated with changes in genes encoding tubulins, the structural units of MTs, MT-associated proteins (MAPs), or additional factors such as MT modifying enzymes which modulating tubulin post-translational modifications (PTMs) regulate MT functions and dynamics. Efforts to use MT-targeting therapeutic agents for the treatment of neurodegenerative diseases are underway. Many of these agents have provided several benefits when tested on both in vitro and in vivo neurodegenerative model systems. Currently, the most frequently addressed therapeutic interventions include drugs that modulate MT stability or that target tubulin PTMs, such as tubulin acetylation. The purpose of this review is to provide an update on the relevance of MT dysfunctions to the process of neurodegeneration and briefly discuss advances in the use of MT-targeting drugs for the treatment of neurodegenerative disorders.
Collapse
|
38
|
Noonepalle S, Shen S, Ptáček J, Tavares MT, Zhang G, Stránský J, Pavlíček J, Ferreira GM, Hadley M, Pelaez G, Bařinka C, Kozikowski AP, Villagra A. Rational Design of Suprastat: A Novel Selective Histone Deacetylase 6 Inhibitor with the Ability to Potentiate Immunotherapy in Melanoma Models. J Med Chem 2020; 63:10246-10262. [PMID: 32815366 DOI: 10.1021/acs.jmedchem.0c00567] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Selective inhibition of histone deacetylase 6 (HDAC6) is being recognized as a therapeutic approach for cancers. In this study, we designed a new HDAC6 inhibitor, named Suprastat, using in silico simulations. X-ray crystallography and molecular dynamics simulations provide strong evidence to support the notion that the aminomethyl and hydroxyl groups in the capping group of Suprastat establish significant hydrogen bond interactions, either direct or water-mediated, with residues D460, N530, and S531, which play a vital role in regulating the deacetylase function of the enzyme and which are absent in other isoforms. In vitro characterization of Suprastat demonstrates subnanomolar HDAC6 inhibitory potency and a hundred- to a thousand-fold HDAC6 selectivity over the other HDAC isoforms. In vivo studies reveal that a combination of Suprastat and anti-PD1 immunotherapy enhances antitumor immune response, mediated by a decrease of protumoral M2 macrophages and increased infiltration of antitumor CD8+ effector and memory T-cells.
Collapse
Affiliation(s)
- Satish Noonepalle
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia 20052, United States
| | - Sida Shen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Jakub Ptáček
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Maurício T Tavares
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Guiping Zhang
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Jan Stránský
- Centre of Molecular Structure, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Jiří Pavlíček
- Centre of Molecular Structure, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Glaucio M Ferreira
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Melissa Hadley
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia 20052, United States
| | - Guido Pelaez
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia 20052, United States
| | - Cyril Bařinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | | | - Alejandro Villagra
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia 20052, United States
| |
Collapse
|
39
|
Turro E, Astle WJ, Megy K, Gräf S, Greene D, Shamardina O, Allen HL, Sanchis-Juan A, Frontini M, Thys C, Stephens J, Mapeta R, Burren OS, Downes K, Haimel M, Tuna S, Deevi SVV, Aitman TJ, Bennett DL, Calleja P, Carss K, Caulfield MJ, Chinnery PF, Dixon PH, Gale DP, James R, Koziell A, Laffan MA, Levine AP, Maher ER, Markus HS, Morales J, Morrell NW, Mumford AD, Ormondroyd E, Rankin S, Rendon A, Richardson S, Roberts I, Roy NBA, Saleem MA, Smith KGC, Stark H, Tan RYY, Themistocleous AC, Thrasher AJ, Watkins H, Webster AR, Wilkins MR, Williamson C, Whitworth J, Humphray S, Bentley DR, Kingston N, Walker N, Bradley JR, Ashford S, Penkett CJ, Freson K, Stirrups KE, Raymond FL, Ouwehand WH. Whole-genome sequencing of patients with rare diseases in a national health system. Nature 2020; 583:96-102. [PMID: 32581362 PMCID: PMC7610553 DOI: 10.1038/s41586-020-2434-2] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 05/05/2020] [Indexed: 02/02/2023]
Abstract
Most patients with rare diseases do not receive a molecular diagnosis and the aetiological variants and causative genes for more than half such disorders remain to be discovered1. Here we used whole-genome sequencing (WGS) in a national health system to streamline diagnosis and to discover unknown aetiological variants in the coding and non-coding regions of the genome. We generated WGS data for 13,037 participants, of whom 9,802 had a rare disease, and provided a genetic diagnosis to 1,138 of the 7,065 extensively phenotyped participants. We identified 95 Mendelian associations between genes and rare diseases, of which 11 have been discovered since 2015 and at least 79 are confirmed to be aetiological. By generating WGS data of UK Biobank participants2, we found that rare alleles can explain the presence of some individuals in the tails of a quantitative trait for red blood cells. Finally, we identified four novel non-coding variants that cause disease through the disruption of transcription of ARPC1B, GATA1, LRBA and MPL. Our study demonstrates a synergy by using WGS for diagnosis and aetiological discovery in routine healthcare.
Collapse
Affiliation(s)
- Ernest Turro
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK.
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK.
| | - William J Astle
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Karyn Megy
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Stefan Gräf
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Daniel Greene
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Olga Shamardina
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Hana Lango Allen
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Alba Sanchis-Juan
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- British Heart Foundation Cambridge Centre of Excellence, University of Cambridge, Cambridge, UK
| | - Chantal Thys
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Jonathan Stephens
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Rutendo Mapeta
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Oliver S Burren
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthias Haimel
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Salih Tuna
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Sri V V Deevi
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Timothy J Aitman
- MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London, UK
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - David L Bennett
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
| | - Paul Calleja
- High Performance Computing Service, University of Cambridge, Cambridge, UK
| | - Keren Carss
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Mark J Caulfield
- Genomics England Ltd, London, UK
- William Harvey Research Institute, NIHR Biomedical Research Centre at Barts, Queen Mary University of London, London, UK
| | - Patrick F Chinnery
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, Cambridge Biomedical Campus, Cambridge, UK
| | - Peter H Dixon
- Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| | - Daniel P Gale
- Department of Renal Medicine, University College London, London, UK
- Rare Renal Disease Registry, UK Renal Registry, Bristol, UK
| | - Roger James
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Ania Koziell
- King's College London, London, UK
- Department of Paediatric Nephrology, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - Michael A Laffan
- Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
- Centre for Haematology, Imperial College London, London, UK
| | - Adam P Levine
- Department of Renal Medicine, University College London, London, UK
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
- Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Bristol, UK
| | - Joannella Morales
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Nicholas W Morrell
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrew D Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
- University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Elizabeth Ormondroyd
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- Department of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stuart Rankin
- High Performance Computing Service, University of Cambridge, Cambridge, UK
| | - Augusto Rendon
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Genomics England Ltd, London, UK
| | - Sylvia Richardson
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Irene Roberts
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Paediatrics, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Noemi B A Roy
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Moin A Saleem
- Bristol Renal and Children's Renal Unit, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol Royal Hospital for Children, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Kenneth G C Smith
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Hannah Stark
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Rhea Y Y Tan
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Bristol, UK
| | - Andreas C Themistocleous
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | | | - Hugh Watkins
- Department of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Andrew R Webster
- Moorfields Eye Hospital NHS Trust, London, UK
- UCL Institute of Opthalmology, University College London, London, UK
| | | | - Catherine Williamson
- Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Faculty of Medicine, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - James Whitworth
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
- Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | | | | | - Nathalie Kingston
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Neil Walker
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - John R Bradley
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, UK
- Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Renal Medicine, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sofie Ashford
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Christopher J Penkett
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Kathleen E Stirrups
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - F Lucy Raymond
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK.
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.
- British Heart Foundation Cambridge Centre of Excellence, University of Cambridge, Cambridge, UK.
- Wellcome Sanger Institute, Cambridge, UK.
| |
Collapse
|
40
|
Mazzetti S, De Leonardis M, Gagliardi G, Calogero AM, Basellini MJ, Madaschi L, Costa I, Cacciatore F, Spinello S, Bramerio M, Cilia R, Rolando C, Giaccone G, Pezzoli G, Cappelletti G. Phospho-HDAC6 Gathers Into Protein Aggregates in Parkinson's Disease and Atypical Parkinsonisms. Front Neurosci 2020; 14:624. [PMID: 32655357 PMCID: PMC7324673 DOI: 10.3389/fnins.2020.00624] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 05/19/2020] [Indexed: 01/10/2023] Open
Abstract
HDAC6 is a unique histone deacetylase that targets cytoplasmic non-histone proteins and has a specific ubiquitin-binding activity. Both of these activities are required for HDAC6-mediated formation of aggresomes, which contain misfolded proteins that will ultimately be degraded via autophagy. HDAC6 deacetylase activity is increased following phosphorylation on serine 22 (phospho-HDAC6). In human, HDAC6 localizes in neuronal Lewy bodies in Parkinson’s disease (PD) and in oligodendrocytic Papp–Lantos bodies in multiple system atrophy (MSA). However, the expression of phospho-HDAC6 in post-mortem human brains is currently unexplored. Here, we evaluate and compare the distribution of HDAC6 and its phosphorylated form in human brains obtained from patients affected by three forms of parkinsonism: two synucleinopathies (PD and MSA) and a tauopathy (progressive supranuclear palsy, PSP). We find that both HDAC6 and its phosphorylated form localize with pathological protein aggregates, including α-synuclein-positive Lewy bodies in PD and Papp–Lantos bodies in MSA, and phospho-tau-positive neurofibrillary tangles in PSP. We further find a direct interaction of HDAC6 with α-synuclein with proximity ligation assay (PLA) in neuronal cell of PD patients. Taken together, our findings suggest that both HDAC6 and phospho-HDAC6 regulate the homeostasis of intra-neuronal proteins in parkinsonism.
Collapse
Affiliation(s)
- Samanta Mazzetti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy.,Fondazione Grigioni per il Morbo di Parkinson, Milan, Italy
| | - Mara De Leonardis
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Gloria Gagliardi
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Alessandra Maria Calogero
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy.,Fondazione Grigioni per il Morbo di Parkinson, Milan, Italy
| | | | - Laura Madaschi
- UNITECH NO LIMITS, Università degli Studi di Milano, Milan, Italy
| | - Ilaria Costa
- Imaging TDU, IFOM Foundation, The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Francesca Cacciatore
- Unit of Neuropathology and Neurology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sonia Spinello
- Unit of Neuropathology and Neurology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Manuela Bramerio
- S. C. Divisione Oncologia Falck and S. C. Divisione Anatomia Patologica, Ospedale Niguarda Ca' Granda, Milan, Italy
| | - Roberto Cilia
- Parkinson Institute, ASST "G.Pini-CTO," Milan, Italy
| | - Chiara Rolando
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Giorgio Giaccone
- Unit of Neuropathology and Neurology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Gianni Pezzoli
- Fondazione Grigioni per il Morbo di Parkinson, Milan, Italy.,Parkinson Institute, ASST "G.Pini-CTO," Milan, Italy
| | - Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy.,Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
41
|
Kershaw S, Morgan DJ, Boyd J, Spiller DG, Kitchen G, Zindy E, Iqbal M, Rattray M, Sanderson CM, Brass A, Jorgensen C, Hussell T, Matthews LC, Ray DW. Glucocorticoids rapidly inhibit cell migration through a novel, non-transcriptional HDAC6 pathway. J Cell Sci 2020; 133:jcs242842. [PMID: 32381682 PMCID: PMC7295589 DOI: 10.1242/jcs.242842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids (GCs) act through the glucocorticoid receptor (GR, also known as NR3C1) to regulate immunity, energy metabolism and tissue repair. Upon ligand binding, activated GR mediates cellular effects by regulating gene expression, but some GR effects can occur rapidly without new transcription. Here, we show that GCs rapidly inhibit cell migration, in response to both GR agonist and antagonist ligand binding. The inhibitory effect on migration is prevented by GR knockdown with siRNA, confirming GR specificity, but not by actinomycin D treatment, suggesting a non-transcriptional mechanism. We identified a rapid onset increase in microtubule polymerisation following GC treatment, identifying cytoskeletal stabilisation as the likely mechanism of action. HDAC6 overexpression, but not knockdown of αTAT1, rescued the GC effect, implicating HDAC6 as the GR effector. Consistent with this hypothesis, ligand-dependent cytoplasmic interaction between GR and HDAC6 was demonstrated by quantitative imaging. Taken together, we propose that activated GR inhibits HDAC6 function, and thereby increases the stability of the microtubule network to reduce cell motility. We therefore report a novel, non-transcriptional mechanism whereby GCs impair cell motility through inhibition of HDAC6 and rapid reorganization of the cell architecture.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Stephen Kershaw
- Systems Oncology, Cancer Research UK Manchester Institute, Manchester, SK10 4TG, UK
| | - David J Morgan
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology and Inflammation University of Manchester, Manchester, M13 9PT, UK
| | - James Boyd
- Division of Cellular and Molecular Physiology, University of Liverpool, Liverpool, L69 3BX, UK
| | - David G Spiller
- Platform Sciences, Enabling Technologies, and Infrastructure, University of Manchester, Manchester, M13 9PT, UK
| | - Gareth Kitchen
- Division of Diabetes, Endocrinology, and Gastroenterology, University of Manchester, Manchester, M13 9PT, UK
| | - Egor Zindy
- Division of Informatics, Imaging, and Data Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Mudassar Iqbal
- Division of Informatics, Imaging, and Data Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Magnus Rattray
- Division of Informatics, Imaging, and Data Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Christopher M Sanderson
- Division of Cellular and Molecular Physiology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Andrew Brass
- Division of Informatics, Imaging, and Data Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Claus Jorgensen
- Systems Oncology, Cancer Research UK Manchester Institute, Manchester, SK10 4TG, UK
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, M13 9PT, UK
- Lydia Becker Institute of Immunology and Inflammation University of Manchester, Manchester, M13 9PT, UK
| | - Laura C Matthews
- Leeds Institute of Cancer and Pathology, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - David W Ray
- Division of Diabetes, Endocrinology, and Gastroenterology, University of Manchester, Manchester, M13 9PT, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, OX3 7LE, and NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| |
Collapse
|
42
|
Vališ K, Novák P. Targeting ERK-Hippo Interplay in Cancer Therapy. Int J Mol Sci 2020; 21:ijms21093236. [PMID: 32375238 PMCID: PMC7247570 DOI: 10.3390/ijms21093236] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular signal-regulated kinase (ERK) is a part of the mitogen-activated protein kinase (MAPK) signaling pathway which allows the transduction of various cellular signals to final effectors and regulation of elementary cellular processes. Deregulation of the MAPK signaling occurs under many pathological conditions including neurodegenerative disorders, metabolic syndromes and cancers. Targeted inhibition of individual kinases of the MAPK signaling pathway using synthetic compounds represents a promising way to effective anti-cancer therapy. Cross-talk of the MAPK signaling pathway with other proteins and signaling pathways have a crucial impact on clinical outcomes of targeted therapies and plays important role during development of drug resistance in cancers. We discuss cross-talk of the MAPK/ERK signaling pathway with other signaling pathways, in particular interplay with the Hippo/MST pathway. We demonstrate the mechanism of cell death induction shared between MAPK/ERK and Hippo/MST signaling pathways and discuss the potential of combination targeting of these pathways in the development of more effective anti-cancer therapies.
Collapse
Affiliation(s)
- Karel Vališ
- Correspondence: (K.V.); (P.N.); Tel.: +420-325873610 (P.N.)
| | - Petr Novák
- Correspondence: (K.V.); (P.N.); Tel.: +420-325873610 (P.N.)
| |
Collapse
|
43
|
Much More Than a Scaffold: Cytoskeletal Proteins in Neurological Disorders. Cells 2020; 9:cells9020358. [PMID: 32033020 PMCID: PMC7072452 DOI: 10.3390/cells9020358] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/08/2023] Open
Abstract
Recent observations related to the structure of the cytoskeleton in neurons and novel cytoskeletal abnormalities involved in the pathophysiology of some neurological diseases are changing our view on the function of the cytoskeletal proteins in the nervous system. These efforts allow a better understanding of the molecular mechanisms underlying neurological diseases and allow us to see beyond our current knowledge for the development of new treatments. The neuronal cytoskeleton can be described as an organelle formed by the three-dimensional lattice of the three main families of filaments: actin filaments, microtubules, and neurofilaments. This organelle organizes well-defined structures within neurons (cell bodies and axons), which allow their proper development and function through life. Here, we will provide an overview of both the basic and novel concepts related to those cytoskeletal proteins, which are emerging as potential targets in the study of the pathophysiological mechanisms underlying neurological disorders.
Collapse
|
44
|
Ustinova K, Novakova Z, Saito M, Meleshin M, Mikesova J, Kutil Z, Baranova P, Havlinova B, Schutkowski M, Matthias P, Barinka C. The disordered N-terminus of HDAC6 is a microtubule-binding domain critical for efficient tubulin deacetylation. J Biol Chem 2020; 295:2614-2628. [PMID: 31953325 DOI: 10.1074/jbc.ra119.011243] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/14/2020] [Indexed: 11/06/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) is a multidomain cytosolic enzyme having tubulin deacetylase activity that has been unequivocally assigned to the second of the tandem catalytic domains. However, virtually no information exists on the contribution of other HDAC6 domains on tubulin recognition. Here, using recombinant protein expression, site-directed mutagenesis, fluorimetric and biochemical assays, microscale thermophoresis, and total internal reflection fluorescence microscopy, we identified the N-terminal, disordered region of HDAC6 as a microtubule-binding domain and functionally characterized it to the single-molecule level. We show that the microtubule-binding motif spans two positively charged patches comprising residues Lys-32 to Lys-58. We found that HDAC6-microtubule interactions are entirely independent of the catalytic domains and are mediated by ionic interactions with the negatively charged microtubule surface. Importantly, a crosstalk between the microtubule-binding domain and the deacetylase domain was critical for recognition and efficient deacetylation of free tubulin dimers both in vitro and in vivo Overall, our results reveal that recognition of substrates by HDAC6 is more complex than previously appreciated and that domains outside the tandem catalytic core are essential for proficient substrate deacetylation.
Collapse
Affiliation(s)
- Kseniya Ustinova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic; Department of Biochemistry, Faculty of Natural Science, Charles University, Albertov 6, Prague 2, Czech Republic
| | - Zora Novakova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Makoto Saito
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; Faculty of Sciences, University of Basel, 4031 Basel, Switzerland
| | - Marat Meleshin
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Charles Tanford Protein Center, Martin Luther University, Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Jana Mikesova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Zsofia Kutil
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Petra Baranova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Barbora Havlinova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Mike Schutkowski
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Charles Tanford Protein Center, Martin Luther University, Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Patrick Matthias
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; Faculty of Sciences, University of Basel, 4031 Basel, Switzerland
| | - Cyril Barinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic.
| |
Collapse
|
45
|
Zessin M, Kutil Z, Meleshin M, Nováková Z, Ghazy E, Kalbas D, Marek M, Romier C, Sippl W, Bařinka C, Schutkowski M. One-Atom Substitution Enables Direct and Continuous Monitoring of Histone Deacylase Activity. Biochemistry 2019; 58:4777-4789. [PMID: 31682411 DOI: 10.1021/acs.biochem.9b00786] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We developed a one-step direct assay for the determination of histone deacylase (HDAC) activity by substituting the carbonyl oxygen of the acyl moiety with sulfur, resulting in thioacylated lysine side chains. This modification is recognized by class I HDACs with different efficiencies ranging from not accepted for HDAC1 to kinetic constants similar to that of the parent oxo substrate for HDAC8. Class II HDACs can hydrolyze thioacylated substrates with approximately 5-10-fold reduced kcat values, which resembles the effect of thioamide substitution in metallo-protease substrates. Class IV HDAC11 accepts thiomyristoyl modification less efficiently with an ∼5-fold reduced specificity constant. On the basis of the unique spectroscopic properties of thioamide bonds (strong absorption in spectral range of 260-280 nm and efficient fluorescence quenching), HDAC-mediated cleavage of thioamides could be followed by ultraviolet-visible and fluorescence spectroscopy in a continuous manner. The HDAC activity assay is compatible with microtiter plate-based screening formats up to 1536-well plates with Z' factors of >0.75 and signal-to-noise ratios of >50. Using thioacylated lysine residues in p53-derived peptides, we optimized substrates for HDAC8 with a catalytic efficiency of >250000 M-1 s-1, which are more than 100-fold more effective than most of the known substrates. We determined inhibition constants of several inhibitors for human HDACs using thioacylated peptidic substrates and found good correlation with the values from the literature. On the other hand, we could introduce N-methylated, N-acylated lysine residues as inhibitors for HDACs with an IC50 value of 1 μM for an N-methylated, N-myristoylated peptide derivative and human HDAC11.
Collapse
Affiliation(s)
- Matthes Zessin
- Department of Medicinal Chemistry, Institute of Pharmacy , Martin-Luther-University Halle-Wittenberg , 06120 Halle/Saale , Germany
| | - Zsófia Kutil
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV , Prumyslova 595 , 252 50 Vestec , Czech Republic
| | - Marat Meleshin
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Charles-Tanford-Protein Center , Martin-Luther-University Halle-Wittenberg , 06120 Halle/Saale , Germany
| | - Zora Nováková
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV , Prumyslova 595 , 252 50 Vestec , Czech Republic
| | - Ehab Ghazy
- Department of Medicinal Chemistry, Institute of Pharmacy , Martin-Luther-University Halle-Wittenberg , 06120 Halle/Saale , Germany
| | - Diana Kalbas
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Charles-Tanford-Protein Center , Martin-Luther-University Halle-Wittenberg , 06120 Halle/Saale , Germany
| | - Martin Marek
- Departement de Biologie Structurale Integrative, Institut de Genetique et Biologie Moleculaire et Cellulaire (IGBMC) , Universite de Strasbourg (UDS), CNRS, INSERM , 1 rue Laurent Fries, B.P. 10142 , 67404 Illkirch Cedex IGBMC, France
| | - Christophe Romier
- Departement de Biologie Structurale Integrative, Institut de Genetique et Biologie Moleculaire et Cellulaire (IGBMC) , Universite de Strasbourg (UDS), CNRS, INSERM , 1 rue Laurent Fries, B.P. 10142 , 67404 Illkirch Cedex IGBMC, France
| | - Wolfgang Sippl
- Department of Medicinal Chemistry, Institute of Pharmacy , Martin-Luther-University Halle-Wittenberg , 06120 Halle/Saale , Germany
| | - Cyril Bařinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV , Prumyslova 595 , 252 50 Vestec , Czech Republic
| | - Mike Schutkowski
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Charles-Tanford-Protein Center , Martin-Luther-University Halle-Wittenberg , 06120 Halle/Saale , Germany
| |
Collapse
|
46
|
Interactions between two regulatory proteins of microtubule dynamics, HDAC6, TPPP/p25, and the hub protein, DYNLL/LC8. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118556. [PMID: 31505170 DOI: 10.1016/j.bbamcr.2019.118556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022]
Abstract
Degradation of unwanted proteins is important in protein quality control cooperating with the dynein/dynactin-mediated trafficking along the acetylated microtubule (MT) network. Proteins associated directly/indirectly with tubulin/MTs play crucial roles in both physiological and pathological processes. Our studies focus on the interrelationship of the tubulin deacetylase HDAC6, the MT-associated TPPP/p25 with its deacetylase inhibitory potency and the hub dynein light chain DYNLL/LC8, constituent of dynein and numerous other protein complexes. In this paper, evidence is provided for the direct interaction of DYNLL/LC8 with TPPP/p25 and HDAC6 and their assembly into binary/ternary complexes with functional potency. The in vitro binding data was obtained with recombinant proteins and used for mathematical modelling. These data and visualization of their localizations by bimolecular fluorescence complementation technology and immunofluorescence microscopy in HeLa cells revealed the promoting effect of TPPP/p25 on the interaction of DYNLL/LC8 with both tubulin and HDAC6. Localization of the LC8-2-TPPP/p25 complex was observed on the MT network in contrast to the LC8-2-HDAC6 complex, which was partly translocated to the nucleus. LC8-2 did not influence directly the acetylation of the MT network. However, the binding of TPPP/p25 to a new binding site of DYNLL/LC8, outside the canonical binding groove, counteracted the TPPP/p25-derived hyperacetylation of the MT network. Our data suggest that multiple associations of the regulatory proteins of the MT network could ensure fine tuning in the regulation of the intracellular trafficking process either by the complexation of DYNLL/LC8 with new partners or indirectly by the modulation of the acetylation level of the MT network.
Collapse
|
47
|
Shen S, Hadley M, Ustinova K, Pavlicek J, Knox T, Noonepalle S, Tavares MT, Zimprich CA, Zhang G, Robers MB, Bařinka C, Kozikowski AP, Villagra A. Discovery of a New Isoxazole-3-hydroxamate-Based Histone Deacetylase 6 Inhibitor SS-208 with Antitumor Activity in Syngeneic Melanoma Mouse Models. J Med Chem 2019; 62:8557-8577. [PMID: 31414801 DOI: 10.1021/acs.jmedchem.9b00946] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Isoxazole is a five-membered heterocycle that is widely used in drug discovery endeavors. Here, we report the design, synthesis, and structural and biological characterization of SS-208, a novel HDAC6-selective inhibitor containing the isoxazole-3-hydroxamate moiety as a zinc-binding group as well as a hydrophobic linker. A crystal structure of the Danio rerio HDAC6/SS-208 complex reveals a bidentate coordination of the active-site zinc ion that differs from the preferred monodentate coordination observed for HDAC6 complexes with phenylhydroxamate-based inhibitors. While SS-208 has minimal effects on the viability of murine SM1 melanoma cells in vitro, it significantly reduced in vivo tumor growth in a murine SM1 syngeneic melanoma mouse model. These findings suggest that the antitumor activity of SS-208 is mainly mediated by immune-related antitumor activity as evidenced by the increased infiltration of CD8+ and NK+ T cells and the enhanced ratio of M1 and M2 macrophages in the tumor microenvironment.
Collapse
Affiliation(s)
- Sida Shen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , University of Illinois at Chicago , Chicago , Illinois 60612 , United States
| | - Melissa Hadley
- Department of Biochemistry and Molecular Medicine , The George Washington University , Washington, District of Columbia 20052 , United States
| | - Kseniya Ustinova
- Laboratory of Structural Biology , Institute of Biotechnology of the Czech Academy of Sciences , Prumyslova 595 , 252 50 Vestec , Czech Republic.,Department of Biochemistry, Faculty of Natural Science , Charles University , Albertov 6 , 128 43 Prague 2 , Czech Republic
| | - Jiri Pavlicek
- Laboratory of Structural Biology , Institute of Biotechnology of the Czech Academy of Sciences , Prumyslova 595 , 252 50 Vestec , Czech Republic
| | - Tessa Knox
- Department of Biochemistry and Molecular Medicine , The George Washington University , Washington, District of Columbia 20052 , United States
| | - Satish Noonepalle
- Department of Biochemistry and Molecular Medicine , The George Washington University , Washington, District of Columbia 20052 , United States
| | - Mauricio T Tavares
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy , University of Illinois at Chicago , Chicago , Illinois 60612 , United States
| | - Chad A Zimprich
- Promega Corporation , Madison , Wisconsin 53711 , United States
| | - Guiping Zhang
- Bontac Bio-Engineering (Shenzhen) Co., Ltd , Shenzhen , Guangdong 518102 , China
| | | | - Cyril Bařinka
- Laboratory of Structural Biology , Institute of Biotechnology of the Czech Academy of Sciences , Prumyslova 595 , 252 50 Vestec , Czech Republic
| | - Alan P Kozikowski
- StarWise Therapeutics LLC, University Research Park, Inc. , Madison , Wisconsin 53719 , United States
| | - Alejandro Villagra
- Department of Biochemistry and Molecular Medicine , The George Washington University , Washington, District of Columbia 20052 , United States
| |
Collapse
|
48
|
The histone deacetylase class I, II inhibitor trichostatin A delays peripheral neurodegeneration. J Mol Histol 2019; 50:167-178. [PMID: 30671879 DOI: 10.1007/s10735-019-09815-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/14/2019] [Indexed: 01/04/2023]
Abstract
Peripheral nerves, which consist of an axon and a unique glial cell called a Schwann cell, transduce signals from the brain and spinal cord to target organs. Peripheral nerve degeneration leads to distal motor or sensory disorders such as diabetic neuropathy, Charcot-Marie-Tooth disease, and Gullain-Barré syndrome, with symptoms such as dysesthesia, speech impairment, vision change, erectile dysfunction, and urinary incontinence. Schwann cells play an important role in peripheral nerve degeneration. Therefore, revealing the characteristics of Schwann cells will be essential in understanding peripheral neurodegeneration-related diseases for which there is currently no effective treatment. Trichostatin A (TSA) is a noncompetitive, reversible inhibitor of class I and II histone deacetylases (HDACs). HDACs have been shown not only to deacetylate histones but also to target non-histone proteins involved in diverse signaling pathways. Recent studies have revealed that diverse HDAC subtypes regulate peripheral neurodegeneration. Thus, regulating HDAC levels could be an effective strategy for the development of drugs targeting peripheral nerve-related diseases. In fact, the use of TSA has been investigated for the treatment of many diseases, including degenerative diseases of the central nervous system; however, the effects of TSA on peripheral neurodegeneration have not yet been well established. In this study, we revealed the effect of TSA on the process of peripheral neurodegeneration. TSA successfully inhibited myelin fragmentation, axonal degradation, and trans-dedifferentiation and proliferation of Schwann cells, which are essential phenotypes in peripheral neurodegeneration. Therefore, TSA could be a potential drug for patients suffering from peripheral neurodegeneration-related diseases.
Collapse
|
49
|
Ortiz Flores RM, Distel JS, Aguilera MO, Berón W. The role of microtubules and the dynein/dynactin motor complex of host cells in the biogenesis of the Coxiella burnetii-containing vacuole. PLoS One 2019; 14:e0209820. [PMID: 30640917 PMCID: PMC6331085 DOI: 10.1371/journal.pone.0209820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 12/12/2018] [Indexed: 01/20/2023] Open
Abstract
Microtubules (Mts) are dynamic cytoskeleton structures that play a key role in vesicular transport. The Mts-mediated transport depends on motor proteins named kinesins and the dynein/dynactin motor complex. The Rab7 adapter protein FYCO1 controls the anterograde transport of the endocytic compartments through the interaction with the kinesin KIF5. Rab7 and its partner RILP induce the recruitment of dynein/dynactin to late endosomes regulating its retrograde transport to the perinuclear area to fuse with lysosomes. The late endosomal-lysosomal fusion is regulated by the HOPS complex through its interaction with RILP and the GTPase Arl8. Coxiella burnetii (Cb), the causative agent of Q fever, is an obligate intracellular pathogen, which generates a large compartment with autophagolysosomal characteristics named Cb-containing vacuole (CCV). The CCV forms through homotypic fusion between small non-replicative CCVs (nrCCV) and through heterotypic fusion with other compartments, such as endosomes and lysosomes. In this work, we characterise the role of Mts, motor proteins, RILP/Rab7 and Arl8 on the CCV biogenesis. The formation of the CCV was affected when either the dynamics and/or the acetylation state of Mts were modified. Similarly, the overexpression of the dynactin subunit non-functional mutants p150Glued and RILP led to the formation of small nrCCVs. This phenomenon is not observed in cells overexpressing WT proteins, the motor KIF5 or its interacting protein FYCO1. The formation of the CCV was normal in infected cells that overexpressed Arl8 alone or together with hVps41 (a HOPS subunit) or in cells co-overexpressing hVps41 and RILP. The dominant negative mutant of Arl8 and the non-functional hVps41 inhibited the formation of the CCV. When the formation of CCV was affected, the bacterial multiplication diminished. Our results suggest that nrCCVs recruit the molecular machinery that regulate the Mts-dependent retrograde transport, Rab7/RILP and the dynein/dynactin system, as well as the tethering processes such as HOPS complex and Arl8 to finally originate the CCV where C. burnetii multiplies.
Collapse
Affiliation(s)
- Rodolfo M. Ortiz Flores
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, Argentina
| | - Jesús S. Distel
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, Argentina
| | - Milton O. Aguilera
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, Argentina
| | - Walter Berón
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, Argentina
- * E-mail:
| |
Collapse
|
50
|
Ruzic D, Petkovic M, Agbaba D, Ganesan A, Nikolic K. Combined Ligand and Fragment‐based Drug Design of Selective Histone Deacetylase – 6 Inhibitors. Mol Inform 2019; 38:e1800083. [DOI: 10.1002/minf.201800083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/08/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| | - Milos Petkovic
- Department of Organic Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| | - Danica Agbaba
- Department of Pharmaceutical Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| | - A. Ganesan
- School of PharmacyUniversity of East Anglia Norwich Research Park NR4 7TJ Norwich United Kingdom
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
- Department of Pharmaceutical Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| |
Collapse
|