151
|
Messaoudi K, Ali A, Ishaq R, Palazzo A, Sliwa D, Bluteau O, Souquère S, Muller D, Diop KM, Rameau P, Lapierre V, Marolleau JP, Matthias P, Godin I, Pierron G, Thomas SG, Watson SP, Droin N, Vainchenker W, Plo I, Raslova H, Debili N. Critical role of the HDAC6-cortactin axis in human megakaryocyte maturation leading to a proplatelet-formation defect. Nat Commun 2017; 8:1786. [PMID: 29176689 PMCID: PMC5702605 DOI: 10.1038/s41467-017-01690-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/10/2017] [Indexed: 01/08/2023] Open
Abstract
Thrombocytopenia is a major side effect of a new class of anticancer agents that target histone deacetylase (HDAC). Their mechanism is poorly understood. Here, we show that HDAC6 inhibition and genetic knockdown lead to a strong decrease in human proplatelet formation (PPF). Unexpectedly, HDAC6 inhibition-induced tubulin hyperacetylation has no effect on PPF. The PPF decrease induced by HDAC6 inhibition is related to cortactin (CTTN) hyperacetylation associated with actin disorganization inducing important changes in the distribution of megakaryocyte (MK) organelles. CTTN silencing in human MKs phenocopies HDAC6 inactivation and knockdown leads to a strong PPF defect. This is rescued by forced expression of a deacetylated CTTN mimetic. Unexpectedly, unlike human-derived MKs, HDAC6 and CTTN are shown to be dispensable for mouse PPF in vitro and platelet production in vivo. Our results highlight an unexpected function of HDAC6–CTTN axis as a positive regulator of human but not mouse MK maturation. Histone deacetylase (HDAC) inhibitors, a class of cancer therapeutics, cause thrombocytopenia via an unknown mechanism. Here, the authors show that HDAC6 inhibition impairs proplatelet formation in human megakaryocytes, and show that this is linked to hyperacetylation of the actin-binding protein cortactin.
Collapse
Affiliation(s)
- Kahia Messaoudi
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France.,Paris7 Diderot University, 75013, Paris, France
| | - Ashfaq Ali
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France.,Paris7 Diderot University, 75013, Paris, France
| | - Rameez Ishaq
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France.,Paris7 Diderot University, 75013, Paris, France
| | - Alberta Palazzo
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France.,Paris7 Diderot University, 75013, Paris, France
| | - Dominika Sliwa
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France
| | - Olivier Bluteau
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France
| | - Sylvie Souquère
- CNRS-UMR-9196, Institut Gustave Roussy, 94805, Villejuif, France
| | - Delphine Muller
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France
| | - Khadija M Diop
- Genomic Platform, Institut Gustave Roussy, 94805, Villejuif, France
| | - Philippe Rameau
- Gustave Roussy, Integrated Biology Core Facility, 94805, Villejuif, France
| | | | - Jean-Pierre Marolleau
- Clinical Hematology and Cell Therapy Department, Amiens Hospital, UPJV University EA4666, 80054, Amiens, France
| | - Patrick Matthias
- Friedrich Miescher Institute for Biomedical Research, 4002, Basel, Switzerland
| | - Isabelle Godin
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France
| | - Gérard Pierron
- CNRS-UMR-9196, Institut Gustave Roussy, 94805, Villejuif, France
| | - Steven G Thomas
- Institute of Cardiovascular Sciences, The Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands, UK
| | - Stephen P Watson
- Institute of Cardiovascular Sciences, The Medical School, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands, UK
| | - Nathalie Droin
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France.,Genomic Platform, Institut Gustave Roussy, 94805, Villejuif, France
| | - William Vainchenker
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France
| | - Isabelle Plo
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France
| | - Hana Raslova
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France.,Paris-Saclay University, UMR 1170, 94805, Villejuif, France.,Gustave Roussy, 94805, Villejuif, France
| | - Najet Debili
- Institut National de la Santé et de la Recherche Médicale, UMR 1170, Equipe labellisée par la Ligue Nationale contre le Cancer, 94805, Villejuif, France. .,Paris-Saclay University, UMR 1170, 94805, Villejuif, France. .,Gustave Roussy, 94805, Villejuif, France.
| |
Collapse
|
152
|
Lernoux M, Schnekenburger M, Dicato M, Diederich M. Anti-cancer effects of naturally derived compounds targeting histone deacetylase 6-related pathways. Pharmacol Res 2017; 129:337-356. [PMID: 29133216 DOI: 10.1016/j.phrs.2017.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/02/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022]
Abstract
Alterations of the epigenetic machinery, affecting multiple biological functions, represent a major hallmark enabling the development of tumors. Among epigenetic regulatory proteins, histone deacetylase (HDAC)6 has emerged as an interesting potential therapeutic target towards a variety of diseases including cancer. Accordingly, this isoenzyme regulates many vital cellular regulatory processes and pathways essential to physiological homeostasis, as well as tumor multistep transformation involving initiation, promotion, progression and metastasis. In this review, we will consequently discuss the critical implications of HDAC6 in distinct mechanisms relevant to physiological and cancerous conditions, as well as the anticancer properties of synthetic, natural and natural-derived compounds through the modulation of HDAC6-related pathways.
Collapse
Affiliation(s)
- Manon Lernoux
- Laboratory of Molecular and Cellular Biology of Cancer, Kirchberg Hospital, 9, Edward Steichen Street, L-2540 Luxembourg, Luxembourg
| | - Michael Schnekenburger
- Laboratory of Molecular and Cellular Biology of Cancer, Kirchberg Hospital, 9, Edward Steichen Street, L-2540 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratory of Molecular and Cellular Biology of Cancer, Kirchberg Hospital, 9, Edward Steichen Street, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, 08826, South Korea.
| |
Collapse
|
153
|
Radomski N, Rebbig A, Leonhardt RM, Knittler MR. Xenophagic pathways and their bacterial subversion in cellular self-defense - παντα ρει - everything is in flux. Int J Med Microbiol 2017; 308:185-196. [PMID: 29126745 DOI: 10.1016/j.ijmm.2017.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/24/2017] [Accepted: 10/29/2017] [Indexed: 01/09/2023] Open
Abstract
Autophagy is an evolutionarily ancient and highly conserved eukaryotic mechanism that targets cytoplasmic material for degradation. Autophagic flux involves the formation of autophagosomes and their degradation by lysosomes. The process plays a crucial role in maintaining cellular homeostasis and responds to various environmental conditions. While autophagy had previously been thought to be a non-selective process, it is now clear that it can also selectively target cellular organelles, such as mitochondria (referred to as mitophagy) and/or invading pathogens (referred to as xenophagy). Selective autophagy is characterized by specific substrate recognition and requires distinct cellular adaptor proteins. Here we review xenophagic mechanisms involved in the recognition and autolysosomal or autophagolysosomal degradation of different intracellular bacteria. In this context, we also discuss a recently discovered cellular self-defense pathway, termed mito-xenophagy, which occurs during bacterial infection of dendritic cells and depends on a TNF-α-mediated metabolic switch from oxidative phosphorylation to glycolysis.
Collapse
Affiliation(s)
- Nadine Radomski
- Institute of Immunology, Friedrich-Loeffler-Institut, Institute of Immunology, Federal Research Institute of Animal Health, D-17493 Greifswald, Isle of Riems, Germany
| | - Annica Rebbig
- Institute of Immunology, Friedrich-Loeffler-Institut, Institute of Immunology, Federal Research Institute of Animal Health, D-17493 Greifswald, Isle of Riems, Germany
| | - Ralf M Leonhardt
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Michael R Knittler
- Institute of Immunology, Friedrich-Loeffler-Institut, Institute of Immunology, Federal Research Institute of Animal Health, D-17493 Greifswald, Isle of Riems, Germany.
| |
Collapse
|
154
|
Harding RJ, Ferreira de Freitas R, Collins P, Franzoni I, Ravichandran M, Ouyang H, Juarez-Ornelas KA, Lautens M, Schapira M, von Delft F, Santhakumar V, Arrowsmith CH. Small Molecule Antagonists of the Interaction between the Histone Deacetylase 6 Zinc-Finger Domain and Ubiquitin. J Med Chem 2017; 60:9090-9096. [DOI: 10.1021/acs.jmedchem.7b00933] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Rachel J. Harding
- Structural
Genomics Consortium, University of Toronto, MaRS South Tower, Suite 700, 101
College Street, Toronto, Ontario M5G 1L7, Canada
| | - Renato Ferreira de Freitas
- Structural
Genomics Consortium, University of Toronto, MaRS South Tower, Suite 700, 101
College Street, Toronto, Ontario M5G 1L7, Canada
| | - Patrick Collins
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot OX11 0QX, U.K
| | - Ivan Franzoni
- Department
of Chemistry, Davenport Chemical Laboratories, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Mani Ravichandran
- Structural
Genomics Consortium, University of Toronto, MaRS South Tower, Suite 700, 101
College Street, Toronto, Ontario M5G 1L7, Canada
| | - Hui Ouyang
- GrandPharma, Wuhan City Plaza, 23rd Floor, 160
Qiaokou Road, Wuhan, Hubei 430032, China
| | - Kevin A. Juarez-Ornelas
- Department
of Chemistry, Davenport Chemical Laboratories, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Mark Lautens
- Department
of Chemistry, Davenport Chemical Laboratories, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| | - Matthieu Schapira
- Structural
Genomics Consortium, University of Toronto, MaRS South Tower, Suite 700, 101
College Street, Toronto, Ontario M5G 1L7, Canada
- Department
of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Frank von Delft
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot OX11 0QX, U.K
- Structural
Genomics Consortium, University of Oxford, Roosevelt Drive, Oxford OX11 9HP, U.K
| | - Vjayaratnam Santhakumar
- Structural
Genomics Consortium, University of Toronto, MaRS South Tower, Suite 700, 101
College Street, Toronto, Ontario M5G 1L7, Canada
| | - Cheryl H. Arrowsmith
- Structural
Genomics Consortium, University of Toronto, MaRS South Tower, Suite 700, 101
College Street, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
155
|
Wloga D, Joachimiak E, Fabczak H. Tubulin Post-Translational Modifications and Microtubule Dynamics. Int J Mol Sci 2017; 18:ijms18102207. [PMID: 29065455 PMCID: PMC5666887 DOI: 10.3390/ijms18102207] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/12/2017] [Accepted: 10/19/2017] [Indexed: 11/24/2022] Open
Abstract
Microtubules are hollow tube-like polymeric structures composed of α,β-tubulin heterodimers. They play an important role in numerous cellular processes, including intracellular transport, cell motility and segregation of the chromosomes during cell division. Moreover, microtubule doublets or triplets form a scaffold of a cilium, centriole and basal body, respectively. To perform such diverse functions microtubules have to differ in their properties. Post-translational modifications are one of the factors that affect the properties of the tubulin polymer. Here we focus on the direct and indirect effects of post-translational modifications of tubulin on microtubule dynamics.
Collapse
Affiliation(s)
- Dorota Wloga
- Laboratory of Cytoskeleton and Cilia Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Str., 02-093 Warsaw, Poland.
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Str., 02-093 Warsaw, Poland.
| | - Hanna Fabczak
- Laboratory of Cytoskeleton and Cilia Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Str., 02-093 Warsaw, Poland.
| |
Collapse
|
156
|
Dubois F, Alpha K, Turner CE. Paxillin regulates cell polarization and anterograde vesicle trafficking during cell migration. Mol Biol Cell 2017; 28:3815-3831. [PMID: 29046398 PMCID: PMC5739297 DOI: 10.1091/mbc.e17-08-0488] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/26/2017] [Accepted: 10/13/2017] [Indexed: 12/25/2022] Open
Abstract
Cell polarization and directed migration play pivotal roles in diverse physiological and pathological processes. Herein, we identify new roles for paxillin-mediated HDAC6 inhibition in regulating key aspects of cell polarization in both two-dimensional and one-dimensional matrix environments. Paxillin, by modulating microtubule acetylation through HDAC6 regulation, was shown to control centrosome and Golgi reorientation toward the leading edge, a hallmark of cell polarization to ensure directed trafficking of promigratory factors. Paxillin was also required for pericentrosomal Golgi localization and centrosome cohesion, independent of its localization to, and role in, focal adhesion signaling. In addition, we provide evidence of an accumulation of paxillin at the centrosome that is dependent on focal adhesion kinase (FAK) and identify an important collaboration between paxillin and FAK signaling in the modulation of microtubule acetylation, as well as centrosome and Golgi organization and polarization. Finally, paxillin was also shown to be required for optimal anterograde vesicular trafficking to the plasma membrane.
Collapse
Affiliation(s)
- Fatemeh Dubois
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Kyle Alpha
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
157
|
Recent advances in the discovery of potent and selective HDAC6 inhibitors. Eur J Med Chem 2017; 143:1406-1418. [PMID: 29133060 DOI: 10.1016/j.ejmech.2017.10.040] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/14/2017] [Accepted: 10/14/2017] [Indexed: 01/07/2023]
Abstract
Histone deacetylase HDAC6, a member of the class IIb HDAC family, is unique among HDAC enzymes in having two active catalytic domains, and has unique physiological function. In addition to the modification of histone, HDAC6 targets specific substrates including α-tubulin and HSP90, and are involved in protein trafficking and degradation, cell shape and migration. Selective HDAC6 inhibitors are an emerging class of pharmaceuticals due to the involvement of HDAC6 in different pathways related to neurodegenerative diseases, cancer, and immunology. Therefore, extensive investigations have been made in the discovery of selective HDAC6 inhibitors. Based on their different zinc binding groups (ZBGs), in this review, HDAC6 inhibitors are grouped as hydroxamic acids, a sulfur containing ZBG based derivatives and other ZBG-derived compounds, and their enzymatic inhibitory activity, selectivity and other biological activities are introduced and summarized.
Collapse
|
158
|
HDAC6 Suppresses Age-Dependent Ectopic Fat Accumulation by Maintaining the Proteostasis of PLIN2 in Drosophila. Dev Cell 2017; 43:99-111.e5. [DOI: 10.1016/j.devcel.2017.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 05/01/2017] [Accepted: 08/31/2017] [Indexed: 01/05/2023]
|
159
|
Fukuda T, Yanagi S. Psychiatric behaviors associated with cytoskeletal defects in radial neuronal migration. Cell Mol Life Sci 2017; 74:3533-3552. [PMID: 28516224 PMCID: PMC11107632 DOI: 10.1007/s00018-017-2539-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/21/2017] [Accepted: 05/11/2017] [Indexed: 12/17/2022]
Abstract
Normal development of the cerebral cortex is an important process for higher brain functions, such as language, and cognitive and social functions. Psychiatric disorders, such as schizophrenia and autism, are thought to develop owing to various dysfunctions occurring during the development of the cerebral cortex. Radial neuronal migration in the embryonic cerebral cortex is a complex process, which is achieved by strict control of cytoskeletal dynamics, and impairments in this process are suggested to cause various psychiatric disorders. Our recent findings indicate that radial neuronal migration as well as psychiatric behaviors is rescued by controlling microtubule stability during the embryonic stage. In this review, we outline the relationship between psychiatric disorders, such as schizophrenia and autism, and radial neuronal migration in the cerebral cortex by focusing on the cytoskeleton and centrosomes. New treatment strategies for psychiatric disorders will be discussed.
Collapse
Affiliation(s)
- Toshifumi Fukuda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan.
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan.
| |
Collapse
|
160
|
Blixt NC, Faulkner BK, Astleford K, Lelich R, Schering J, Spencer E, Gopalakrishnan R, Jensen ED, Mansky KC. Class II and IV HDACs function as inhibitors of osteoclast differentiation. PLoS One 2017; 12:e0185441. [PMID: 28953929 PMCID: PMC5617211 DOI: 10.1371/journal.pone.0185441] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023] Open
Abstract
Histone deacetylases (HDACs) are negative regulators of transcription and have been shown to regulate specific changes in gene expression. In vertebrates, eighteen HDACs have thus far been identified and subdivided into four classes (I-IV). Key roles for several HDACs in bone development and biology have been elucidated through in vitro and in vivo models. By comparison, there is a paucity of data on the roles of individual HDACs in osteoclast formation and function. In this study, we investigated the gene expression patterns and the effects of suppressing individual class II (Hdac4, 5, 6, 9, and 10) and class IV (Hdac11) HDACs during osteoclast differentiation. We demonstrated that HDAC class II and IV members are differentially expressed during osteoclast differentiation. Additionally, individual shRNA-mediated suppression of Hdac4, 5, 9, 10 and 11 expression resulted in increased multinucleated osteoclast size and demineralization activity, with little to no change in the overall number of multinucleated osteoclasts formed compared with control shRNA-treated cells. We also detected increased expression of genes highly expressed in osteoclasts, including c-Fos, Nfatc1, Dc-stamp and Cathepsin K. These observations indicate that HDACs cooperatively regulate shared targets in a non-redundant manner.
Collapse
Affiliation(s)
- Nicholas C. Blixt
- Departmment of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bora K. Faulkner
- Departmment of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Kristina Astleford
- Department of Developmental and Surgical Sciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Rosemary Lelich
- Department of Developmental and Surgical Sciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jacob Schering
- Department of Developmental and Surgical Sciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ekaterina Spencer
- Department of Developmental and Surgical Sciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Rajaram Gopalakrishnan
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Eric D. Jensen
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail: (EDJ); (KCM)
| | - Kim C. Mansky
- Department of Developmental and Surgical Sciences, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail: (EDJ); (KCM)
| |
Collapse
|
161
|
Human histone deacetylase 6 shows strong preference for tubulin dimers over assembled microtubules. Sci Rep 2017; 7:11547. [PMID: 28912522 PMCID: PMC5599508 DOI: 10.1038/s41598-017-11739-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/29/2017] [Indexed: 12/31/2022] Open
Abstract
Human histone deacetylase 6 (HDAC6) is the major deacetylase responsible for removing the acetyl group from Lys40 of α-tubulin (αK40), which is located lumenally in polymerized microtubules. Here, we provide a detailed kinetic analysis of tubulin deacetylation and HDAC6/microtubule interactions using individual purified components. Our data unequivocally show that free tubulin dimers represent the preferred HDAC6 substrate, with a K M value of 0.23 µM and a deacetylation rate over 1,500-fold higher than that of assembled microtubules. We attribute the lower deacetylation rate of microtubules to both longitudinal and lateral lattice interactions within tubulin polymers. Using TIRF microscopy, we directly visualized stochastic binding of HDAC6 to assembled microtubules without any detectable preferential binding to microtubule tips. Likewise, indirect immunofluorescence microscopy revealed that microtubule deacetylation by HDAC6 is carried out stochastically along the whole microtubule length, rather than from the open extremities. Our data thus complement prior studies on tubulin acetylation and further strengthen the rationale for the correlation between tubulin acetylation and microtubule age.
Collapse
|
162
|
Borgas D, Chambers E, Newton J, Ko J, Rivera S, Rounds S, Lu Q. Cigarette Smoke Disrupted Lung Endothelial Barrier Integrity and Increased Susceptibility to Acute Lung Injury via Histone Deacetylase 6. Am J Respir Cell Mol Biol 2017; 54:683-96. [PMID: 26452072 DOI: 10.1165/rcmb.2015-0149oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Epidemiologic evidence indicates that cigarette smoke (CS) is associated with the development of acute lung injury (ALI). We have previously shown that brief CS exposure exacerbates lipopolysaccharide (LPS)-induced ALI in vivo and endothelial barrier dysfunction in vitro. In this study, we found that CS also exacerbated Pseudomonas-induced ALI in mice. We demonstrated that lung microvascular endothelial cells (ECs) isolated from mice exposed to CS had a greater permeability or incomplete recovery after challenges by LPS and thrombin. Histone deacetylase (HDAC) 6 deacetylates proteins essential for maintenance of endothelial barrier function. We found that HDAC6 phosphorylation at serine-22 was increased in lung tissues of mice exposed to CS and in lung ECs exposed to cigarette smoke extract (CSE). Inhibition of HDAC6 attenuated CSE-induced increases in EC permeability and CS priming of ALI. Similar barrier protection was provided by the microtubule stabilizer taxol, which preserved α-tubulin acetylation. CSE decreased α-tubulin acetylation and caused microtubule depolymerization. In coordination with increased HDAC6 phosphorylation, CSE inhibited Akt and activated glycogen synthase kinase (GSK)-3β; these effects were ameliorated by the antioxidant N-acetyl cysteine. Our results suggest that CS increases lung EC permeability, thereby enhancing susceptibility to ALI, likely through oxidative stress-induced Akt inactivation and subsequent GSK-3β activation. Activated GSK-3β may activate HDAC6 via phosphorylation of serine-22, leading to α-tubulin deacetylation and microtubule disassembly. Inhibition of HDAC6 may be a novel therapeutic option for ALI in cigarette smokers.
Collapse
Affiliation(s)
- Diana Borgas
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Eboni Chambers
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Julie Newton
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Junsuk Ko
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Stephanie Rivera
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
163
|
Abstract
Chlamydiae are bacterial pathogens that grow in vacuolar inclusions. Dendritic cells (DCs) disintegrate these compartments, thereby eliminating the microbes, through auto/xenophagy, which also promotes chlamydial antigen presentation via MHC I. Here, we show that TNF-α controls this pathway by driving cytosolic phospholipase (cPLA)2-mediated arachidonic acid (AA) production. AA then impairs mitochondrial function, which disturbs the development and integrity of these energy-dependent parasitic inclusions, while a simultaneous metabolic switch towards aerobic glycolysis promotes DC survival. Tubulin deacetylase/autophagy regulator HDAC6 associates with disintegrated inclusions, thereby further disrupting their subcellular localisation and stability. Bacterial remnants are decorated with defective mitochondria, mito-aggresomal structures, and components of the ubiquitin/autophagy machinery before they are degraded via mito-xenophagy. The mechanism depends on cytoprotective HSP25/27, the E3 ubiquitin ligase Parkin and HDAC6 and promotes chlamydial antigen generation for presentation on MHC I. We propose that this novel mito-xenophagic pathway linking innate and adaptive immunity is critical for effective DC-mediated anti-bacterial resistance.
Collapse
|
164
|
Novel Class IIa-Selective Histone Deacetylase Inhibitors Discovered Using an in Silico Virtual Screening Approach. Sci Rep 2017; 7:3228. [PMID: 28607401 PMCID: PMC5468338 DOI: 10.1038/s41598-017-03417-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/27/2017] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylases (HDAC) contain eighteen isoforms that can be divided into four classes. Of these isoform enzymes, class IIa (containing HDAC4, 5, 7 and 9) target unique substrates, some of which are client proteins associated with epigenetic control. Class IIa HDACs are reportedly associated with some neuronal disorders, making HDACs therapeutic targets for treating neurodegenerative diseases. Additionally, some reported HDAC inhibitors contain hydroxamate moiety that chelates with zinc ion to become the cofactor of HDAC enzymes. However, the hydroxamate functional group is shown to cause undesirable effects and has poor pharmacokinetic profile. This study used in silico virtual screening methodology to identify several nonhydroxamate compounds, obtained from National Cancer Institute database, which potentially inhibited HDAC4. Comparisons of the enzyme inhibitory activity against a panel of HDAC isoforms revealed these compounds had strong inhibitory activity against class IIa HDACs, but weak inhibitory activity against class I HDACs. Further analysis revealed that a single residue affects the cavity size between class I and class IIa HDACs, thus contributing to the selectivity of HDAC inhibitors discovered in this study. The discovery of these inhibitors presents the possibility of developing new therapeutic treatments that can circumvent the problems seen in traditional hydroxamate-based drugs.
Collapse
|
165
|
Noritsugu K, Ito A, Nakao Y, Yoshida M. Identification of zinc finger transcription factor EGR2 as a novel acetylated protein. Biochem Biophys Res Commun 2017; 489:455-459. [PMID: 28576496 DOI: 10.1016/j.bbrc.2017.05.170] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 05/29/2017] [Indexed: 01/05/2023]
Abstract
EGR2 is a zinc finger transcription factor that regulates myelination in the peripheral nervous system and T cell anergy. The transcriptional activity of EGR2 is known to be regulated by its co-activators and/or co-repressors. Although the activity of transcription factors is generally regulated not only by interactions with co-regulators but also posttranslational modifications including acetylation, little is known about posttranslational modifications of EGR2. Here we show that EGR2 is a novel acetylated protein. Through immunoblotting analyses using an antibody that specifically recognizes the acetylated form of EGR2, CBP and p300 were identified as acetyltransferases, while HDAC6, 10 and SIRT1 were identified as deacetylases of EGR2. Although the NuRD complex containing HDAC1 and HDAC2 is known to associate with EGR2, the present study suggests that acetylation of EGR2 is regulated independently of NuRD.
Collapse
Affiliation(s)
- Kota Noritsugu
- Chemical Genetics Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Department of Biotechnology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Akihiro Ito
- Chemical Genetics Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| | - Yoichi Nakao
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Minoru Yoshida
- Chemical Genetics Laboratory, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Department of Biotechnology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
166
|
Traoré MM, Zwick V, Simões-Pires CA, Nurisso A, Issa M, Cuendet M, Maynadier M, Wein S, Vial H, Jamet H, Wong YS. Hydroxyl Ketone-Based Histone Deacetylase Inhibitors To Gain Insight into Class I HDAC Selectivity versus That of HDAC6. ACS OMEGA 2017; 2:1550-1562. [PMID: 30023639 PMCID: PMC6044785 DOI: 10.1021/acsomega.6b00481] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/29/2017] [Indexed: 06/07/2023]
Abstract
Little is known about the biological and structural features that govern the isoform selectivity for class I histone deacetylases (HDACs) over HDAC6. In addition to that for known inhibitors, like benzamides, psammaplin A, and cyclodepsipeptide-derived thiols, selectivity was also observed for naturally occurring cyclopeptide HDAC inhibitors with an aliphatic flexible linker and ketonelike zinc-binding group (ZBG). The present study reports that this isoform selectivity is mainly due to the linker and ZBG, as replacement of the cyclopeptide cap region by a simple aniline retained class I HDAC isoform selectivity toward HDAC6 in enzymatic assays. The best cyclopeptide-free analogues preserved efficacy against Plasmodium falciparum and cancer cell lines. Molecular modeling provided hypotheses to explain this selectivity and suggests different behaviors of the flexible linker on HDAC1 and HDAC6 pockets, which may influence, on the basis of the strength of the ZBG, its coordination with the zinc ion.
Collapse
Affiliation(s)
- Mohamed
D. M. Traoré
- Département
de Pharmacochimie Moléculaire, CNRS
UMR 5063, ICMG FR 2607, Univ. Grenoble Alpes, 470 rue de la chimie, 38041 Grenoble cedex 9, France
- Département
de Chimie Moléculaire, CNRS UMR 5250,
ICMG FR 2607, Univ. Grenoble Alpes, 301 rue de la chimie, 38041 Grenoble cedex 9, France
| | - Vincent Zwick
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
| | - Claudia A. Simões-Pires
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
| | - Alessandra Nurisso
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
- Laboratoire
Dynamique des Interactions Membranaires Normales et Pathologiques, UMR 5235, CNRS, University of Montpellier, Place Eugène Bataillon, 34095 Montpellier, France
| | - Mark Issa
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
| | - Muriel Cuendet
- School
of Pharmaceutical Sciences, University of
Geneva, University of Lausanne, rue Michel Servet 1, 1211 Geneva, Switzerland
| | - Marjorie Maynadier
- Département
de Biochimie, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Sharon Wein
- Département
de Biochimie, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Henri Vial
- Département
de Biochimie, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Helene Jamet
- Département
de Chimie Moléculaire, CNRS UMR 5250,
ICMG FR 2607, Univ. Grenoble Alpes, 301 rue de la chimie, 38041 Grenoble cedex 9, France
| | - Yung-Sing Wong
- Département
de Pharmacochimie Moléculaire, CNRS
UMR 5063, ICMG FR 2607, Univ. Grenoble Alpes, 470 rue de la chimie, 38041 Grenoble cedex 9, France
| |
Collapse
|
167
|
Zan J, Liu S, Sun DN, Mo KK, Yan Y, Liu J, Hu BL, Gu JY, Liao M, Zhou JY. Rabies Virus Infection Induces Microtubule Depolymerization to Facilitate Viral RNA Synthesis by Upregulating HDAC6. Front Cell Infect Microbiol 2017; 7:146. [PMID: 28491824 PMCID: PMC5405082 DOI: 10.3389/fcimb.2017.00146] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 04/07/2017] [Indexed: 12/20/2022] Open
Abstract
Rabies virus (RABV) is the cause of rabies, and is associated with severe neurological symptoms, high mortality rate, and a serious threat to human health. Although cellular tubulin has recently been identified to be incorporated into RABV particles, the effects of RABV infection on the microtubule cytoskeleton remain poorly understood. In this study, we show that RABV infection induces microtubule depolymerization as observed by confocal microscopy, which is closely associated with the formation of the filamentous network of the RABV M protein. Depolymerization of microtubules significantly increases viral RNA synthesis, while the polymerization of microtubules notably inhibits viral RNA synthesis and prevents the viral M protein from inducing the formation of the filamentous network. Furthermore, the histone deacetylase 6 (HDAC6) expression level progressively increases during RABV infection, and the inhibition of HDAC6 deacetylase activity significantly decreases viral RNA synthesis. In addition, the expression of viral M protein alone was found to significantly upregulate HDAC6 expression, leading to a substantial reduction in its substrate, acetylated α-tubulin, eventually resulting in microtubule depolymerization. These results demonstrate that HDAC6 plays a positive role in viral transcription and replication by inducing microtubule depolymerization during RABV infection.
Collapse
Affiliation(s)
- Jie Zan
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang UniversityHangzhou, China
| | - Song Liu
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang UniversityHangzhou, China
| | - Dong-Nan Sun
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang UniversityHangzhou, China
| | - Kai-Kun Mo
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang UniversityHangzhou, China
| | - Yan Yan
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang UniversityHangzhou, China
| | - Juan Liu
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang UniversityHangzhou, China
| | - Bo-Li Hu
- Institute of Immunology, Nanjing Agricultural UniversityNanjing, China
| | - Jin-Yan Gu
- Collaborative Innovation Center and State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang UniversityHangzhou, China
| | - Min Liao
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang UniversityHangzhou, China
| | - Ji-Yong Zhou
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang UniversityHangzhou, China.,Collaborative Innovation Center and State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang UniversityHangzhou, China
| |
Collapse
|
168
|
The therapeutic hope for HDAC6 inhibitors in malignancy and chronic disease. Clin Sci (Lond) 2017; 130:987-1003. [PMID: 27154743 DOI: 10.1042/cs20160084] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/01/2016] [Indexed: 12/12/2022]
Abstract
Recent years have witnessed an emergence of a new class of therapeutic agents, termed histone deacetylase 6 (HDAC6) inhibitors. HDAC6 is one isoform of a family of HDAC enzymes that catalyse the removal of functional acetyl groups from proteins. It stands out from its cousins in almost exclusively deacetylating cytoplasmic proteins, in exerting deacetylation-independent effects and in the success that has been achieved in developing relatively isoform-specific inhibitors of its enzymatic action that have reached clinical trial. HDAC6 plays a pivotal role in the removal of misfolded proteins and it is this role that has been most successfully targeted to date. HDAC6 inhibitors are being investigated for use in combination with proteasome inhibitors for the treatment of lymphoid malignancies, whereby HDAC6-dependent protein disposal currently limits the cytotoxic effectiveness of the latter. Similarly, numerous recent studies have linked altered HDAC6 activity to the pathogenesis of neurodegenerative diseases that are characterized by misfolded protein accumulation. It seems likely though that the function of HDAC6 is not limited to malignancy and neurodegeneration, the deacetylase being implicated in a number of other cellular processes and diseases including in cardiovascular disease, inflammation, renal fibrosis and cystogenesis. Here, we review the unique features of HDAC6 that make it so appealing as a drug target and its currently understood role in health and disease. Whether HDAC6 inhibition will ultimately find a clinical niche in the treatment of malignancy or prevalent complex chronic diseases remains to be determined.
Collapse
|
169
|
Schatten H, Sun QY. Centrosome and microtubule functions and dysfunctions in meiosis: implications for age-related infertility and developmental disorders. Reprod Fertil Dev 2017; 27:934-43. [PMID: 25903261 DOI: 10.1071/rd14493] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/18/2015] [Indexed: 12/13/2022] Open
Abstract
The effects of oocyte aging on meiotic spindle dynamics have been well recognised, but the mechanisms underlying the effects are not well understood. In this paper we review the role of centrosomes and the microtubule cytoskeleton in meiotic spindle formation and maintenance, and the impact of oocyte aging on spindle integrity resulting in centrosome and microtubule dysfunctions that are associated with aneuploidy. Loss of spindle integrity includes dispersion of proteins from the centrosome core structure and loss of attachment of microtubules to centrosomes and kinetochores, which will result in abnormal chromosome separation. The inability of centrosomal proteins to accurately associate with the centrosome structure may be the result of destabilisation of the core structure itself or of microtubule destabilisation at the centrosome-facing microtubule areas that are acetylated in fresh oocytes but may not be acetylated in aging oocytes. Microtubule destabilisation prevents accurate motor-driven transport of centrosomal proteins along microtubules to form and maintain a functional centrosome. Other factors to form and maintain the MII spindle include signal transductions that affect microtubule dynamics and stability. Understanding the mechanisms underlying centrosome and microtubule dysfunctions during oocyte aging will allow diagnosis and analysis of oocyte quality and abnormalities as important aspects for targeted treatment of aging oocytes to extend or restore viability and developmental capacity. New therapeutic approaches will allow improvements in reproductive success rates in IVF clinics, as well as improvements in reproductive success rates in farm animals. This review is focused on: (1) centrosome and microtubule dynamics in fresh and aging oocytes; (2) regulation of centrosome and/or microtubule dynamics and function; and (3) possible treatments to extend the oocyte's reproductive capacity and viability span.
Collapse
Affiliation(s)
- Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, 1600 E Rollins Street, Columbia, MO 65211, USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China
| |
Collapse
|
170
|
Avdoshina V, Caragher SP, Wenzel ED, Taraballi F, Mocchetti I, Harry GJ. The viral protein gp120 decreases the acetylation of neuronal tubulin: potential mechanism of neurotoxicity. J Neurochem 2017; 141:606-613. [PMID: 28295345 DOI: 10.1111/jnc.14015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/10/2017] [Accepted: 02/28/2017] [Indexed: 01/18/2023]
Abstract
The human immunodeficiency virus (HIV) envelope protein gp120 promotes axonal damage and neurite pruning, similar to that observed in HIV-positive subjects with neurocognitive disorders. Thus, gp120 has been used to examine molecular and cellular pathways underlying HIV-mediated neuronal dysfunction. Gp120 binds to tubulin beta III, a component of neuronal microtubules. Microtubule function, which modulates the homeostasis of neurons, is regulated by polymerization and post-translational modifications. Based on these considerations, we tested the hypothesis that gp120 induces dynamic instability of neuronal microtubules. We first observed that gp120 prevents the normal polymerization of tubulin in vitro. We then tested whether gp120 alters the post-translational modifications in tubulin by examining the ability of gp120 to change the levels of acetylated tubulin in primary rat neuronal cultures. Gp120 elicited a time-dependent decrease in tubulin acetylation that was reversed by Helix-A peptide, a compound that competitively displaces the binding of gp120 to neuronal microtubules. To determine whether post-translational modifications in tubulin also occur in vivo, we measured acetylated tubulin in the cerebral cortex of HIV transgenic rats (HIV-tg). We observed a decrease in tubulin acetylation in 5- and 9-month-old HIV-tg rats when compared to age-matched wild type. Neither changes in microglia morphology nor alterations in mRNA levels for interleukin-1β and tumor necrosis factor α were detected in 5-month-old animals. Our findings propose neuronal microtubule instability as a novel mechanism of HIV neurotoxicity, without evidence of enhanced inflammation.
Collapse
Affiliation(s)
- Valeria Avdoshina
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Washington, Washington, District of Columbia, USA
| | - Seamus P Caragher
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Washington, Washington, District of Columbia, USA
| | - Erin D Wenzel
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Washington, Washington, District of Columbia, USA.,Department of Pharmacology and Physiology, Georgetown University Washington, Washington, District of Columbia, USA
| | - Francesca Taraballi
- Department of Regenerative Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Italo Mocchetti
- Laboratory of Preclinical Neurobiology, Department of Neuroscience, Georgetown University Washington, Washington, District of Columbia, USA
| | - Gaylia Jean Harry
- National Toxicology Program Laboratory, National Institute of Environmental Health Science, Research Triangle Park, North Carolina, USA
| |
Collapse
|
171
|
Zheng K, Jiang Y, He Z, Kitazato K, Wang Y. Cellular defence or viral assist: the dilemma of HDAC6. J Gen Virol 2017; 98:322-337. [PMID: 27959772 DOI: 10.1099/jgv.0.000679] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) is a unique cytoplasmic deacetylase that regulates various important biological processes by preventing protein aggregation and deacetylating different non-histone substrates including tubulin, heat shock protein 90, cortactin, retinoic acid inducible gene I and β-catenin. Growing evidence has indicated a dual role for HDAC6 in viral infection and pathogenesis: HDAC6 may represent a host defence mechanism against viral infection by modulating microtubule acetylation, triggering antiviral immune response and stimulating protective autophagy, or it may be hijacked by the virus to enhance proinflammatory response. In this review, we will highlight current data illustrating the complexity and importance of HDAC6 in viral pathogenesis. We will summarize the structure and functional specificity of HDAC6, and its deacetylase- and ubiquitin-dependent activity in key cellular events in response to virus infection. We will also discuss how HDAC6 exerts its direct or indirect histone modification ability in viral lytic-latency switch.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Pharmacy, School of Medicine, Shenzhen University, Shenzhen 518060, PR China.,College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| | - Yingchun Jiang
- Department of Pharmacy, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Zhendan He
- Department of Pharmacy, School of Medicine, Shenzhen University, Shenzhen 518060, PR China
| | - Kaio Kitazato
- Division of Molecular Pharmacology of Infectious Agents, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Yifei Wang
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou 510632, PR China
| |
Collapse
|
172
|
Fütterer A, de Celis J, Navajas R, Almonacid L, Gutiérrez J, Talavera-Gutiérrez A, Pacios-Bras C, Bernascone I, Martin-Belmonte F, Martinéz-A C. DIDO as a Switchboard that Regulates Self-Renewal and Differentiation in Embryonic Stem Cells. Stem Cell Reports 2017; 8:1062-1075. [PMID: 28330622 PMCID: PMC5390109 DOI: 10.1016/j.stemcr.2017.02.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 12/28/2022] Open
Abstract
Transition from symmetric to asymmetric cell division requires precise coordination of differential gene expression. We show that embryonic stem cells (ESCs) mainly express DIDO3 and that their differentiation after leukemia inhibitory factor withdrawal requires DIDO1 expression. C-terminal truncation of DIDO3 (Dido3ΔCT) impedes ESC differentiation while retaining self-renewal; small hairpin RNA-Dido1 ESCs have the same phenotype. Dido3ΔCT ESC differentiation is rescued by ectopic expression of DIDO3, which binds the Dido locus via H3K4me3 and RNA POL II and induces DIDO1 expression. DIDO1, which is exported to cytoplasm, associates with, and is N-terminally phosphorylated by PKCiota. It binds the E3 ubiquitin ligase WWP2, which contributes to cell fate by OCT4 degradation, to allow expression of primitive endoderm (PE) markers. PE formation also depends on phosphorylated DIDO3 localization to centrosomes, which ensures their correct positioning for PE cell polarization. We propose that DIDO isoforms act as a switchboard that regulates genetic programs for ESC transition from pluripotency maintenance to promotion of differentiation. DIDO3 regulates DIDO1 expression Cytoplasmic DIDO1 promotes cell fate DIDO3 regulates centrosome position DIDO1 and DIDO3 are phosphorylated by PKCiota
Collapse
Affiliation(s)
- Agnes Fütterer
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Jésus de Celis
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Rosana Navajas
- Proteomics Unit, ProteoRed ISCIII, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Luis Almonacid
- Genomics Unit, Q-PCR Facility, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Julio Gutiérrez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Amaia Talavera-Gutiérrez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Cristina Pacios-Bras
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Ilenia Bernascone
- Department of Development and Differentiation, Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Fernando Martin-Belmonte
- Department of Development and Differentiation, Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Carlos Martinéz-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain.
| |
Collapse
|
173
|
Li X, Mei Y, Yan B, Vitriol E, Huang S, Ji P, Qiu Y. Histone deacetylase 6 regulates cytokinesis and erythrocyte enucleation through deacetylation of formin protein mDia2. Haematologica 2017; 102:984-994. [PMID: 28255013 PMCID: PMC5451330 DOI: 10.3324/haematol.2016.161513] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/27/2017] [Indexed: 11/09/2022] Open
Abstract
The formin protein mDia2 plays a critical role in a number of cellular processes through its ability to promote nucleation and elongation of actin filaments. In erythroblasts, this includes control of cytokinesis and enucleation by regulating contractile actin ring formation. Here we report a novel mechanism of how mDia2 is regulated: through acetylation and deacetylation at lysine 970 in the formin homology 2 domain. Ectopic expression of an acetyl-mimic mDia2 mutant in mouse erythroblasts is sufficient to abolish contractile actin ring formation at the cleavage furrow and subsequent erythrocyte cytokinesis and enucleation. We also identified that class II histone deacetylase 6 deacetylates and subsequently activates mDia2. Knockdown or inhibition of histone deacetylase 6 impairs contractile actin ring formation, and expression of a non-acetyl-mimic mDia2 mutant restores the contractile actin ring and rescues the impairment of enucleation. In addition to revealing a new step in mDia2 regulation, this study may unveil a novel regulatory mechanism of formin-mediated actin assembly, since the K970 acetylation site is conserved among Dia proteins
Collapse
Affiliation(s)
- Xuehui Li
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yang Mei
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Bowen Yan
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Eric Vitriol
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Suming Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA.,Macau Institute for Applied Research in Medicine and Health, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Peng Ji
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yi Qiu
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
174
|
Choi H, Kim HJ, Kim J, Kim S, Yang J, Lee W, Park Y, Hyeon SJ, Lee DS, Ryu H, Chung J, Mook-Jung I. Increased acetylation of Peroxiredoxin1 by HDAC6 inhibition leads to recovery of Aβ-induced impaired axonal transport. Mol Neurodegener 2017; 12:23. [PMID: 28241840 PMCID: PMC5330132 DOI: 10.1186/s13024-017-0164-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Background Reduction or inhibition of histone deacetylase 6 (HDAC6) has been shown to rescue memory in mouse models of Alzheimer’s disease (AD) and is recently being considered a possible therapeutic strategy. However, the restoring mechanism of HDAC6 inhibition has not been fully understood. Methods and results Here, we found that an anti-oxidant protein Peroxdiredoxin1 (Prx1), a substrate of HDAC6, malfunctions in Aβ treated cells, the brains of 5xFAD AD model mice and AD patients. Malfunctioning Prx1, caused by reduced Prx1 acetylation levels, was recovered by HDAC6 inhibition. Increasing acetylation levels of Prx1 by HDAC6 inhibition recovered elevated reactive oxygen species (ROS) levels, elevated Ca2+ levels and impaired mitochondrial axonal transport, sequentially, even in the presence of Aβ. Prx1 mutant studies on the K197 site for an acetylation mimic or silencing mutation support the results showing that HDAC6 inhibitor restores Aβ-induced disruption of ROS, Ca2+ and axonal transport. Conclusions Taken together, increasing acetylation of Prx1 by HDAC6 inhibition has several beneficial effects in AD pathology. Here, we present the novel mechanism by which elevated acetylation of Prx1 rescues mitochondrial axonal transport impaired by Aβ. Therefore, our results suggest that modulation of Prx1 acetylation by HDAC6 inhibition has great therapeutic potential for AD and has further therapeutic possibilities for other neurodegenerative diseases as well. Electronic supplementary material The online version of this article (doi:10.1186/s13024-017-0164-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Heesun Choi
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Haeng Jun Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Jisoo Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Soohyun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University, College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jinhee Yang
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Wonik Lee
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea
| | - Yeonju Park
- Department of Biomedical Sciences, Laboratory of Immunology and Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Jae Hyeon
- Center for Neuromedicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Laboratory of Immunology and Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Hoon Ryu
- VA Boston Healthcare System, Boston University Alzheimer's Disease Center, and Department of Neurology, Boston University School of Medicine, Boston, MA02130, USA.,Center for Neuromedicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University, College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Korea.
| |
Collapse
|
175
|
Pelletier N, Grégoire S, Yang XJ. Assays for Acetylation and Other Acylations of Lysine Residues. ACTA ACUST UNITED AC 2017; 87:14.11.1-14.11.18. [PMID: 28150880 DOI: 10.1002/cpps.26] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Lysine acetylation refers to addition of an acetyl moiety to the epsilon-amino group of a lysine residue and is important for regulating protein functions in various organisms from bacteria to humans. This is a reversible and precisely controlled covalent modification that either serves as an on/off switch or participates in a codified manner with other post-translational modifications to regulate different cellular and developmental processes in normal and pathological states. This unit describes methods for in vitro and in vivo determination of lysine acetylation. Such methods can be easily extended for analysis of other acylations (such as propionylation, butyrylation, crotonylation, and succinylation) that are also present in histones and many other proteins. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Nadine Pelletier
- Goodman Cancer Research Center and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Serge Grégoire
- Goodman Cancer Research Center and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Xiang-Jiao Yang
- Goodman Cancer Research Center and Department of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
176
|
Takács Z, Jankovics F, Vilmos P, Lénárt P, Röper K, Erdélyi M. The spectraplakin Short stop is an essential microtubule regulator involved in epithelial closure in Drosophila. J Cell Sci 2017; 130:712-724. [PMID: 28062848 PMCID: PMC5339884 DOI: 10.1242/jcs.193003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 12/21/2016] [Indexed: 02/04/2023] Open
Abstract
Dorsal closure of the Drosophila embryonic epithelium provides an excellent model system for the in vivo analysis of molecular mechanisms regulating cytoskeletal rearrangements. In this study, we investigated the function of the Drosophila spectraplakin Short stop (Shot), a conserved cytoskeletal structural protein, during closure of the dorsal embryonic epithelium. We show that Shot is essential for the efficient final zippering of the opposing epithelial margins. By using isoform-specific mutant alleles and genetic rescue experiments with truncated Shot variants, we demonstrate that Shot functions as an actin-microtubule cross-linker in mediating zippering. At the leading edge of epithelial cells, Shot regulates protrusion dynamics by promoting filopodia formation. Fluorescence recovery after photobleaching (FRAP) analysis and in vivo imaging of microtubule growth revealed that Shot stabilizes dynamic microtubules. The actin- and microtubule-binding activities of Shot are simultaneously required in the same molecule, indicating that Shot is engaged as a physical crosslinker in this process. We propose that Shot-mediated interactions between microtubules and actin filaments facilitate filopodia formation, which promotes zippering by initiating contact between opposing epithelial cells.
Collapse
Affiliation(s)
- Zsanett Takács
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, Szeged 6726, Hungary
| | - Ferenc Jankovics
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, Szeged 6726, Hungary
| | - Péter Vilmos
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, Szeged 6726, Hungary
| | - Péter Lénárt
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Katja Röper
- MRC-Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Miklós Erdélyi
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62, Szeged 6726, Hungary
| |
Collapse
|
177
|
Soci UPR, Melo SFS, Gomes JLP, Silveira AC, Nóbrega C, de Oliveira EM. Exercise Training and Epigenetic Regulation: Multilevel Modification and Regulation of Gene Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:281-322. [PMID: 29098627 DOI: 10.1007/978-981-10-4304-8_16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Exercise training elicits acute and adaptive long term changes in human physiology that mediate the improvement of performance and health state. The responses are integrative and orchestrated by several mechanisms, as gene expression. Gene expression is essential to construct the adaptation of the biological system to exercise training, since there are molecular processes mediating oxidative and non-oxidative metabolism, angiogenesis, cardiac and skeletal myofiber hypertrophy, and other processes that leads to a greater physiological status. Epigenetic is the field that studies about gene expression changes heritable by meiosis and mitosis, by changes in chromatin and DNA conformation, but not in DNA sequence, that studies the regulation on gene expression that is independent of genotype. The field approaches mechanisms of DNA and chromatin conformational changes that inhibit or increase gene expression and determine tissue specific pattern. The three major studied epigenetic mechanisms are DNA methylation, Histone modification, and regulation of noncoding RNA-associated genes. This review elucidates these mechanisms, focusing on the relationship between them and their relationship with exercise training, physical performance and the enhancement of health status. On this chapter, we clarified the relationship of epigenetic modulations and their intimal relationship with acute and chronic effect of exercise training, concentrating our effort on skeletal muscle, heart and vascular responses, that are the most responsive systems against to exercise training and play crucial role on physical performance and improvement of health state.
Collapse
Affiliation(s)
| | | | | | | | - Clara Nóbrega
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, SP, Brazil
| | | |
Collapse
|
178
|
Jeong SG, Cho GW. The tubulin deacetylase sirtuin-2 regulates neuronal differentiation through the ERK/CREB signaling pathway. Biochem Biophys Res Commun 2017; 482:182-187. [DOI: 10.1016/j.bbrc.2016.11.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 12/14/2022]
|
179
|
YOSHIDA M, KUDO N, KOSONO S, ITO A. Chemical and structural biology of protein lysine deacetylases. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:297-321. [PMID: 28496053 PMCID: PMC5489435 DOI: 10.2183/pjab.93.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 03/21/2017] [Indexed: 05/27/2023]
Abstract
Histone acetylation is a reversible posttranslational modification that plays a fundamental role in regulating eukaryotic gene expression and chromatin structure/function. Key enzymes for removing acetyl groups from histones are metal (zinc)-dependent and NAD+-dependent histone deacetylases (HDACs). The molecular function of HDACs have been extensively characterized by various approaches including chemical, molecular, and structural biology, which demonstrated that HDACs regulate cell proliferation, differentiation, and metabolic homeostasis, and that their alterations are deeply involved in various human disorders including cancer. Notably, drug discovery efforts have achieved success in developing HDAC-targeting therapeutics for treatment of several cancers. However, recent advancements in proteomics technology have revealed much broader aspects of HDACs beyond gene expression control. Not only histones but also a large number of cellular proteins are subject to acetylation by histone acetyltransferases (HATs) and deacetylation by HDACs. Furthermore, some of their structures can flexibly accept and hydrolyze other acyl groups on protein lysine residues. This review mainly focuses on structural aspects of HDAC enzymatic activity regulated by interaction with substrates, co-factors, small molecule inhibitors, and activators.
Collapse
Affiliation(s)
- Minoru YOSHIDA
- RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Norio KUDO
- RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Saori KOSONO
- RIKEN Center for Sustainable Resource Science, Saitama, Japan
- Biotechnology Research Center, The University of Tokyo, Tokyo, Japan
| | - Akihiro ITO
- RIKEN Center for Sustainable Resource Science, Saitama, Japan
| |
Collapse
|
180
|
MIIP accelerates epidermal growth factor receptor protein turnover and attenuates proliferation in non-small cell lung cancer. Oncotarget 2016; 7:9118-34. [PMID: 26824318 PMCID: PMC4891030 DOI: 10.18632/oncotarget.7001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 01/17/2016] [Indexed: 12/22/2022] Open
Abstract
The migration and invasion inhibitory protein (MIIP) has been discovered recently to have inhibitory functions in cell proliferation and migration. Overexpression of MIIP reduced the intracellular steady-state level of epidermal growth factor receptor (EGFR) protein in lung cancer cells with no effect on EGFR mRNA expression compared to that in the control cells. This MIIP-promoted EGFR protein degradation was reversed by proteasome and lysosome inhibitors, suggesting the involvement of both proteasomal and lysosomal pathways in this degradation. This finding was further validated by pulse-chase experiments using 35S-methionine metabolic labeling. We found that MIIP accelerates EGFR protein turnover via proteasomal degradation in the endoplasmic reticulum and then via the lysosomal pathway after its entry into endocytic trafficking. MIIP-stimulated downregulation of EGFR inhibits downstream activation of Ras and blocks the MEK signal transduction pathway, resulting in inhibition of cell proliferation. The negative correlation between MIIP and EGFR protein expression was validated in lung adenocarcinoma samples. Furthermore, the higher MIIP protein expression predicts a better overall survival of Stage IA-IIIA lung adenocarcinoma patients who underwent radical surgery. These findings reveal a new mechanism by which MIIP inhibits cell proliferation.
Collapse
|
181
|
Qin HT, Li HQ, Liu F. Selective histone deacetylase small molecule inhibitors: recent progress and perspectives. Expert Opin Ther Pat 2016; 27:621-636. [PMID: 28033734 DOI: 10.1080/13543776.2017.1276565] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Since the first pan-HDAC inhibitor SAHA was approved by U.S. FDA 10 years ago, HDACs including SIRT1-7 have received significant attention due to the fact that aberrant histone deacetylase activtiy has been implicated in a variety of human diseases, such as cancers, virus infection, and neurodegenerative diseases. During the past years, a considerable achievement of development of isoform- or class-selective HDAC inhibitors has been made, yielding many drug candidates for further clinical studies, which represents a state-of-the-art technology in the drug discovery arena. Areas covered: This review covers new patents and articles about isoform- or class-selective HDAC inhibitors during the last four years, as well as the therapeutic potential of these compounds. Expert opinion: HDACs represent one of the most promising therapeutic targets, particularly for tumor therapy though their roles in cancer are still blurry. From 2012 to present, along with the advances of structural biology and homology models, lots of isoform- or class-selective HDAC inhibitors, such as hydroxamic acids and benzamides with various capping groups were found, providing a promising way to circumvent drug toxicity and side-effect issues, as well as providing chemical probes for further better understanding of the biological process related to specific isoform.
Collapse
Affiliation(s)
- Hai-Tao Qin
- a Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Department of Medicinal Chemistry , College of Pharmaceutical Sciences, Soochow University , Suzhou , PR China
| | - Huan-Qiu Li
- b Department of Medicinal Chemistry , College of Pharmaceutical Sciences, Soochow University , Suzhou , PR China
| | - Feng Liu
- a Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Department of Medicinal Chemistry , College of Pharmaceutical Sciences, Soochow University , Suzhou , PR China
| |
Collapse
|
182
|
Dual Chromatin and Cytoskeletal Remodeling by SETD2. Cell 2016; 166:950-962. [PMID: 27518565 DOI: 10.1016/j.cell.2016.07.005] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 05/13/2016] [Accepted: 06/30/2016] [Indexed: 02/06/2023]
Abstract
Posttranslational modifications (PTMs) of tubulin specify microtubules for specialized cellular functions and comprise what is termed a "tubulin code." PTMs of histones comprise an analogous "histone code," although the "readers, writers, and erasers" of the cytoskeleton and epigenome have heretofore been distinct. We show that methylation is a PTM of dynamic microtubules and that the histone methyltransferase SET-domain-containing 2 (SETD2), which is responsible for H3 lysine 36 trimethylation (H3K36me3) of histones, also methylates α-tubulin at lysine 40, the same lysine that is marked by acetylation on microtubules. Methylation of microtubules occurs during mitosis and cytokinesis and can be ablated by SETD2 deletion, which causes mitotic spindle and cytokinesis defects, micronuclei, and polyploidy. These data now identify SETD2 as a dual-function methyltransferase for both chromatin and the cytoskeleton and show a requirement for methylation in maintenance of genomic stability and the integrity of both the tubulin and histone codes.
Collapse
|
183
|
Richter-Landsberg C. Protein aggregate formation in oligodendrocytes: tau and the cytoskeleton at the intersection of neuroprotection and neurodegeneration. Biol Chem 2016; 397:185-94. [PMID: 26083267 DOI: 10.1515/hsz-2015-0157] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/11/2015] [Indexed: 01/01/2023]
Abstract
Oligodendrocytes are dependent on an intact, dynamic microtubule (MT) network, which participates in the elaboration and stabilization of myelin forming extensions, and is essential for cellular sorting processes. The microtubule-associated protein tau is constituent of oligodendrocytes. During culture maturation it is developmentally regulated and important for MT stability, MT formation and intracellular trafficking. Downregulation of tau impairs process outgrowth and the transport of myelin basic protein (MBP) mRNA to the cell periphery. Cells fail to differentiate into MBP-expressing, sheet-forming oligodendrocytes. Tau-positive inclusions originating in oligodendrocytes and white matter pathology are prominent in frontotemporal dementias, such as Pick's disease, progressive supranuclear palsy and corticobasal degeneration. An impairment or overload of the proteolytic degradation systems, i.e. the ubiquitin proteasomal system and the lysosomal degradation pathway, has been connected to the formation of protein aggregates. Large protein aggregates are excluded from the proteasome and degraded by autophagy, which is a highly selective process and requires receptor proteins for ubiquitinated proteins, including histone deacetylase 6 (HDAC6). HDAC6 is present in oligodendrocytes, and α-tubulin and tau are substrates of HDAC6. In this review our current knowledge of the role of tau and protein aggregate formation in oligodendrocyte cell culture systems is summarized.
Collapse
|
184
|
Brill MS, Kleele T, Ruschkies L, Wang M, Marahori NA, Reuter MS, Hausrat TJ, Weigand E, Fisher M, Ahles A, Engelhardt S, Bishop DL, Kneussel M, Misgeld T. Branch-Specific Microtubule Destabilization Mediates Axon Branch Loss during Neuromuscular Synapse Elimination. Neuron 2016; 92:845-856. [PMID: 27773584 PMCID: PMC5133389 DOI: 10.1016/j.neuron.2016.09.049] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 08/14/2016] [Accepted: 09/21/2016] [Indexed: 01/17/2023]
Abstract
Developmental axon remodeling is characterized by the selective removal of branches from axon arbors. The mechanisms that underlie such branch loss are largely unknown. Additionally, how neuronal resources are specifically assigned to the branches of remodeling arbors is not understood. Here we show that axon branch loss at the developing mouse neuromuscular junction is mediated by branch-specific microtubule severing, which results in local disassembly of the microtubule cytoskeleton and loss of axonal transport in branches that will subsequently dismantle. Accordingly, pharmacological microtubule stabilization delays neuromuscular synapse elimination. This branch-specific disassembly of the cytoskeleton appears to be mediated by the microtubule-severing enzyme spastin, which is dysfunctional in some forms of upper motor neuron disease. Our results demonstrate a physiological role for a neurodegeneration-associated modulator of the cytoskeleton, reveal unexpected cell biology of branch-specific axon plasticity and underscore the mechanistic similarities of axon loss in development and disease. During synapse elimination, retreating axon branches dismantle their microtubules Microtubules are destabilized due to branch-specific severing Microtubule stabilization delays axon branch removal during synapse elimination The disease-associated microtubule severing protein spastin mediates microtubule loss
Collapse
Affiliation(s)
- Monika S Brill
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany.
| | - Tatjana Kleele
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Laura Ruschkies
- University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH), Institute for Molecular Neurogenetics, Falkenried 94, 20251 Hamburg, Germany
| | - Mengzhe Wang
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Natalia A Marahori
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Miriam S Reuter
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Torben J Hausrat
- University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH), Institute for Molecular Neurogenetics, Falkenried 94, 20251 Hamburg, Germany
| | - Emily Weigand
- Ball State University, Department of Biology, 2000 West University, Muncie, IN 47306, USA
| | - Matthew Fisher
- Ball State University, Department of Biology, 2000 West University, Muncie, IN 47306, USA
| | - Andrea Ahles
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany; German Center for Cardiovascular Research, DZHK, Partner site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany; German Center for Cardiovascular Research, DZHK, Partner site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Derron L Bishop
- Indiana University School of Medicine, Department of Cellular and Integrative Physiology, Medical Science Building 385, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15(th) Street, Indianapolis, IN 46202, USA
| | - Matthias Kneussel
- University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH), Institute for Molecular Neurogenetics, Falkenried 94, 20251 Hamburg, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany; Center of Integrated Protein Science (CIPSM), Butenandtstraße 5-13, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany.
| |
Collapse
|
185
|
Compagnucci C, Piermarini E, Sferra A, Borghi R, Niceforo A, Petrini S, Piemonte F, Bertini E. Cytoskeletal dynamics during in vitro neurogenesis of induced pluripotent stem cells (iPSCs). Mol Cell Neurosci 2016; 77:113-124. [PMID: 27756615 DOI: 10.1016/j.mcn.2016.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/24/2016] [Accepted: 10/13/2016] [Indexed: 12/27/2022] Open
Abstract
Patient-derived induced pluripotent stem cells (iPSCs) provide a novel tool to investigate the pathophysiology of poorly known diseases, in particular those affecting the nervous system, which has been difficult to study for its lack of accessibility. In this emerging and promising field, recent iPSCs studies are mostly used as "proof-of-principle" experiments that are confirmatory of previous findings obtained from animal models and postmortem human studies; its promise as a discovery tool is just beginning to be realized. A recent number of studies point to the functional similarities between in vitro neurogenesis and in vivo neuronal development, suggesting that similar morphogenetic and patterning events direct neuronal differentiation. In this context, neuronal adhesion, cytoskeletal organization and cell metabolism emerge as an integrated and unexplored processes of human neurogenesis, mediated by the lack of data due to the difficult accessibility of the human neural tissue. These observations raise the necessity to understand which are the players controlling cytoskeletal reorganization and remodeling. In particular, we investigated human in vitro neurogenesis using iPSCs of healthy subjects to unveil the underpinnings of the cytoskeletal dynamics with the aim to shed light on the physiologic events controlling the development and the functionality of neuronal cells. We validate the iPSCs system to better understand the development of the human nervous system in order to set the bases for the future understanding of pathologies including developmental disorders (i.e. intellectual disability), epilepsy but also neurodegenerative disorders (i.e. Friedreich's Ataxia). We investigate the changes of the cytoskeletal components during the 30days of neuronal differentiation and we demonstrate that human neuronal differentiation requires a (time-dependent) reorganization of actin filaments, intermediate filaments and microtubules; and that immature neurons present a finely regulated localization of Glu-, Tyr- and Acet-TUBULINS. This study advances our understanding on cytoskeletal dynamics with the hope to pave the way for future therapies that could be potentially able to target cytoskeletal based neurodevelopmental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Claudia Compagnucci
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Children's Research Hospital Bambino Gesù, IRCCS, Rome 00146, Italy.
| | - Emanuela Piermarini
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Children's Research Hospital Bambino Gesù, IRCCS, Rome 00146, Italy
| | - Antonella Sferra
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Children's Research Hospital Bambino Gesù, IRCCS, Rome 00146, Italy
| | - Rossella Borghi
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Children's Research Hospital Bambino Gesù, IRCCS, Rome 00146, Italy
| | - Alessia Niceforo
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Children's Research Hospital Bambino Gesù, IRCCS, Rome 00146, Italy
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Laboratories, Bambino Gesù Children's Hospital, IRCCS, Rome 00146, Italy
| | - Fiorella Piemonte
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Children's Research Hospital Bambino Gesù, IRCCS, Rome 00146, Italy
| | - Enrico Bertini
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Children's Research Hospital Bambino Gesù, IRCCS, Rome 00146, Italy
| |
Collapse
|
186
|
Deficiency of RITA results in multiple mitotic defects by affecting microtubule dynamics. Oncogene 2016; 36:2146-2159. [DOI: 10.1038/onc.2016.372] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/08/2016] [Accepted: 08/28/2016] [Indexed: 01/31/2023]
|
187
|
Nogueira-Rodrigues J, Brites P, Sousa MM. Axonal pathology in Krabbe's disease: The cytoskeleton as an emerging therapeutic target. J Neurosci Res 2016; 94:1037-41. [DOI: 10.1002/jnr.23771] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/18/2016] [Accepted: 04/29/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Joana Nogueira-Rodrigues
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC) and Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Porto Portugal
| | - Pedro Brites
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC) and Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Porto Portugal
| | - Mónica Mendes Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC) and Instituto de Investigação e Inovação em Saúde; Universidade do Porto; Porto Portugal
| |
Collapse
|
188
|
Desjardins D, Liu Y, Crosson CE, Ablonczy Z. Histone Deacetylase Inhibition Restores Retinal Pigment Epithelium Function in Hyperglycemia. PLoS One 2016; 11:e0162596. [PMID: 27617745 PMCID: PMC5019386 DOI: 10.1371/journal.pone.0162596] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/25/2016] [Indexed: 12/15/2022] Open
Abstract
In diabetic individuals, macular edema is a major cause of vision loss. This condition is refractory to insulin therapy and has been attributed to metabolic memory. The retinal pigment epithelium (RPE) is central to maintaining fluid balance in the retina, and this function is compromised by the activation of advanced glycation end-product receptors (RAGE). Here we provide evidence that acute administration of the RAGE agonist, glycated-albumin (gAlb) or vascular endothelial growth factor (VEGF), increased histone deacetylase (HDAC) activity in RPE cells. The administration of the class I/II HDAC inhibitor, trichostatin-A (TSA), suppressed gAlb-induced reductions in RPE transepithelial resistance (in vitro) and fluid transport (in vivo). Systemic TSA also restored normal RPE fluid transport in rats with subchronic hyperglycemia. Both gAlb and VEGF increased HDAC activity and reduced acetyl-α-tubulin levels. Tubastatin-A, a relatively specific antagonist of HDAC6, inhibited gAlb-induced changes in RPE cell resistance. These data are consistent with the idea that RPE dysfunction following exposure to gAlb, VEGF, or hyperglycemia is associated with increased HDAC6 activity and decreased acetyl-α-tubulin. Therefore, we propose inhibiting HDAC6 in the RPE as a potential therapy for preserving normal fluid homeostasis in the hyperglycemic retina.
Collapse
Affiliation(s)
- Danielle Desjardins
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, 29425, United States of America
| | - Yueying Liu
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, 29425, United States of America
| | - Craig E. Crosson
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, 29425, United States of America
| | - Zsolt Ablonczy
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, 29425, United States of America
- * E-mail:
| |
Collapse
|
189
|
Ren Y, Su X, Kong L, Li M, Zhao X, Yu N, Kang J. Therapeutic effects of histone deacetylase inhibitors in a murine asthma model. Inflamm Res 2016; 65:995-1008. [PMID: 27565183 PMCID: PMC5075027 DOI: 10.1007/s00011-016-0984-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/15/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE AND DESIGN To investigate the therapeutic effects of various HDAC inhibitors on the development of chronic allergic airway disease in mice with airway inflammation, airway remodeling, and airway hyperresponsiveness. SUBJECTS Wild-type BALB/C mice (N = 72). TREATMENT Tubastatin A HCl [TSA, a selective histone deacetylase 6 (HDAC6) inhibitor], PCI-34051 (a selective HDAC8 inhibitor), and givinostat (a broad-spectrum HDAC inhibitor that inhibits class I and class II HDACs and several pro-inflammatory cytokines). METHODS Mice were divided into six groups: control, asthma, dexamethasone (positive control), TSA, PCI-34051, and givinostat (n = 12 per group). Twenty-four hours after OVA nebulization, airway hyperresponsiveness, inflammation, and remodeling were assessed. RESULTS The chronic asthma mouse model produced typical airway inflammation, airway remodeling, and airway hyperresponsiveness. Administration of PCI-34051 and dexamethasone reduced the eosinophilic inflammation and airway hyperresponsiveness in asthma to reduce the airway remodeling. Treatment with Tubastatin A HCl reduced airway inflammation and was associated with decreased IL-4, IL-5 and total inflammatory cell count, as well as goblet cell metaplasia and subepithelial fibrosis; however, this outcome was not as effective as that with dexamethasone. TGF-β1 expression in the cytoplasm of airway epithelium of mice in the Tubastatin A HCl group was reduced and expression of α-SMA in the airway smooth muscle was also decreased. CONCLUSIONS The results suggested that treatment with HDAC inhibitors can reduce airway inflammation, airway remodeling, and airway hyperresponsiveness in chronic allergic airway disease in mice.
Collapse
Affiliation(s)
- Yuan Ren
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, People's Republic of China
| | - Xinming Su
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, People's Republic of China.
| | - Lingfei Kong
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, People's Republic of China
| | - Menglu Li
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, People's Republic of China
| | - Xuan Zhao
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, People's Republic of China
| | - Na Yu
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, People's Republic of China
| | - Jian Kang
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
190
|
Patel H, Stavrou I, Shrestha RL, Draviam V, Frame MC, Brunton VG. Kindlin1 regulates microtubule function to ensure normal mitosis. J Mol Cell Biol 2016; 8:338-48. [PMID: 26993041 PMCID: PMC4991666 DOI: 10.1093/jmcb/mjw009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/13/2015] [Accepted: 11/08/2015] [Indexed: 11/14/2022] Open
Abstract
Loss of Kindlin 1 (Kin1) results in the skin blistering disorder Kindler Syndrome (KS), whose symptoms also include skin atrophy and reduced keratinocyte proliferation. Kin1 binds to integrins to modulate their activation and more recently it has been shown to regulate mitotic spindles and cell survival in a Plk1-dependent manner. Here we report that short-term Kin1 deletion in mouse skin results in impaired mitosis, which is associated with reduced acetylated tubulin (ac-tub) levels and cell proliferation. In cells, impaired mitosis and reduced ac-tub levels are also accompanied by reduced microtubule stability, all of which are rescued by HDAC6 inhibition. The ability of Kin1 to regulate HDAC6-dependent cellular ac-tub levels is dependent on its phosphorylation by Plk1. Taken together, these data define a novel role for Kin1 in microtubule acetylation and stability and offer a mechanistic insight into how certain KS phenotypes, such as skin atrophy and reduced cell proliferation, arise.
Collapse
Affiliation(s)
- Hitesh Patel
- Edinburgh Cancer Research Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Ifigeneia Stavrou
- Edinburgh Cancer Research Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Roshan L Shrestha
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Viji Draviam
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK Present address: School of Biological and Chemical Sciences, Queen Mary University of London, London, E11 4NS, UK
| | - Margaret C Frame
- Edinburgh Cancer Research Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Valerie G Brunton
- Edinburgh Cancer Research Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| |
Collapse
|
191
|
Sasaki N, Nakamura M, Kodama A, Urata Y, Shiokawa N, Hayashi T, Sano A. Chorein interacts with α‐tubulin and histone deacetylase 6, and overexpression preserves cell viability during nutrient deprivation in human embryonic kidney 293 cells. FASEB J 2016; 30:3726-3732. [DOI: 10.1096/fj.201500191rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 07/18/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Natsuki Sasaki
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Masayuki Nakamura
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Akiko Kodama
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Yuka Urata
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Nari Shiokawa
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Takehiro Hayashi
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Akira Sano
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| |
Collapse
|
192
|
Structural insights into HDAC6 tubulin deacetylation and its selective inhibition. Nat Chem Biol 2016; 12:748-54. [DOI: 10.1038/nchembio.2140] [Citation(s) in RCA: 227] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 06/22/2016] [Indexed: 12/31/2022]
|
193
|
The growing landscape of tubulin acetylation: lysine 40 and many more. Biochem J 2016; 473:1859-68. [DOI: 10.1042/bcj20160172] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/29/2016] [Indexed: 11/17/2022]
Abstract
Tubulin heterodimers are the building block of microtubules, which are major elements of the cytoskeleton. Several types of post-translational modifications are found on tubulin subunits as well as on the microtubule polymer to regulate the multiple roles of microtubules. Acetylation of lysine 40 (K40) of the α-tubulin subunit is one of these post-translational modifications which has been extensively studied. We summarize the current knowledge about the structural aspects of K40 acetylation, the functional consequences, the enzymes involved and their regulation. Most importantly, we discuss the potential importance of the recently discovered additional acetylation acceptor lysines in tubulin subunits and highlight the urgent need to study tubulin acetylation in a more integrated perspective.
Collapse
|
194
|
Mitochondrial Metabolism Power SIRT2-Dependent Deficient Traffic Causing Alzheimer’s-Disease Related Pathology. Mol Neurobiol 2016; 54:4021-4040. [DOI: 10.1007/s12035-016-9951-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/06/2016] [Indexed: 01/21/2023]
|
195
|
Skoge RH, Ziegler M. SIRT2 inactivation reveals a subset of hyperacetylated perinuclear microtubules inaccessible to HDAC6. J Cell Sci 2016; 129:2972-82. [PMID: 27311481 DOI: 10.1242/jcs.187518] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/10/2016] [Indexed: 01/07/2023] Open
Abstract
Deacetylation of α-tubulin at lysine 40 is catalyzed by two enzymes, the NAD-dependent deacetylase SIRT2 and the NAD-independent deacetylase HDAC6, in apparently redundant reactions. In the present study, we tested whether these two enzymes might have distinguishable preferences for the deacetylation of different microtubule structures. Using various agents, we induced tubulin hyperacetylation and analyzed the ensuing formation of distinct microtubule structures. HDAC6 inhibition led to general hyperacetylation of the microtubule network throughout the cell, whereas hyperacetylation induced by SIRT2 inactivation was limited to perinuclear microtubules. Hyperacetylation of these perinuclear microtubules was undiminished following HDAC6 overexpression, whereas reactivation of SIRT2 restored the basal acetylation level and a normal microtubule network. By contrast, SIRT2 and HDAC6 acted similarly on the morphologically different, hyperacetylated microtubule structures induced by taxol, MAP2c overexpression or hyperosmotic stress. These results indicate overlapping and distinct functions of HDAC6 and SIRT2. We propose that the differential activity of the two deacetylases, which target the same acetylated lysine residue, might be related to the recognition of specific structural contexts.
Collapse
Affiliation(s)
- Renate Hvidsten Skoge
- Department of Molecular Biology, University of Bergen, Postbox 7803, Bergen 5020, Norway
| | - Mathias Ziegler
- Department of Molecular Biology, University of Bergen, Postbox 7803, Bergen 5020, Norway
| |
Collapse
|
196
|
Yoo J, Kim SJ, Son D, Seo H, Baek SY, Maeng CY, Lee C, Kim IS, Jung YH, Lee SM, Park HJ. Computer-aided identification of new histone deacetylase 6 selective inhibitor with anti-sepsis activity. Eur J Med Chem 2016; 116:126-135. [DOI: 10.1016/j.ejmech.2016.03.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 02/06/2023]
|
197
|
Yu J, Ma Z, Shetty S, Ma M, Fu J. Selective HDAC6 inhibition prevents TNF-α-induced lung endothelial cell barrier disruption and endotoxin-induced pulmonary edema. Am J Physiol Lung Cell Mol Physiol 2016; 311:L39-47. [PMID: 27190059 DOI: 10.1152/ajplung.00051.2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/17/2016] [Indexed: 12/16/2022] Open
Abstract
Lung endothelial damage contributes to the pathogenesis of acute lung injury. New strategies against lung endothelial barrier dysfunction may provide therapeutic benefits against lung vascular injury. Cell-cell junctions and microtubule cytoskeleton are basic components in maintaining endothelial barrier integrity. HDAC6, a deacetylase primarily localized in the cytoplasm, has been reported to modulate nonnuclear protein function through deacetylation. Both α-tubulin and β-catenin are substrates for HDAC6. Here, we examined the effects of tubastatin A, a highly selective HDAC6 inhibitor, on TNF-α induced lung endothelial cell barrier disruption and endotoxin-induced pulmonary edema. Selective HDAC6 inhibition by tubastatin A blocked TNF-α-induced lung endothelial cell hyperpermeability, which was associated with increased α-tubulin acetylation and microtubule stability. Tubastatin A pretreatment inhibited TNF-α-induced endothelial cell contraction and actin stress fiber formation with reduced myosin light chain phosphorylation. Selective HDAC6 inhibition by tubastatin A also induced β-catenin acetylation in human lung endothelial cells, which was associated with increased membrane localization of β-catenin and stabilization of adherens junctions. HDAC6 knockdown by small interfering RNA also prevented TNF-α-induced barrier dysfunction and increased α-tubulin and β-catenin acetylation in endothelial cells. Furthermore, in a mouse model of endotoxemia, tubastatin A was able to prevent endotoxin-induced deacetylation of α-tubulin and β-catenin in lung tissues, which was associated with reduced pulmonary edema. Collectively, our data indicate that selective HDAC6 inhibition by tubastatin A is a potent approach against lung endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Jinyan Yu
- The Second Hospital of Jilin University, Jilin, China; Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, Kentucky; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| | - Zhongsen Ma
- The Second Hospital of Jilin University, Jilin, China
| | - Sreerama Shetty
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas
| | - Mengshi Ma
- The Second Hospital of Jilin University, Jilin, China; Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Jian Fu
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, Kentucky; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| |
Collapse
|
198
|
Yang Y, Zhou J. CYLD - a deubiquitylase that acts to fine-tune microtubule properties and functions. J Cell Sci 2016; 129:2289-95. [PMID: 27173491 DOI: 10.1242/jcs.183319] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Microtubules are dynamic structures that are crucially involved in a variety of cellular activities. The dynamic properties and functions of microtubules are regulated by various factors, such as tubulin isotype composition and microtubule-binding proteins. Initially identified as a deubiquitylase with tumor-suppressing functions, the protein cylindromatosis (CYLD) has recently been revealed to interact with microtubules, modulate microtubule dynamics, and participate in the regulation of cell migration, cell cycle progression, chemotherapeutic drug sensitivity and ciliogenesis. These findings have greatly enriched our understanding of the roles of CYLD in physiological and pathological conditions. Here, we focus on recent literature that shows how CYLD impacts on microtubule properties and functions in various biological processes, and discuss the challenges we face when interpreting results obtained from different experimental systems.
Collapse
Affiliation(s)
- Yunfan Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China Institute of Biomedical Sciences, College of Life Sciences, Key Laboratory of Animal Resistance of Shandong Province, Key Laboratory of Molecular and Nano Probes of the Ministry of Education, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
199
|
Liang Y, Meng D, Zhu B, Pan J. Mechanism of ciliary disassembly. Cell Mol Life Sci 2016; 73:1787-802. [PMID: 26869233 PMCID: PMC11108551 DOI: 10.1007/s00018-016-2148-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 12/19/2022]
Abstract
As motile organelles and sensors, cilia play pivotal roles in cell physiology, development and organ homeostasis. Ciliary defects are associated with a class of cilia-related diseases or developmental disorders, termed ciliopathies. Even though the presence of cilia is required for diverse functions, cilia can be removed through ciliary shortening or resorption that necessitates disassembly of the cilium, which occurs normally during cell cycle progression, cell differentiation and in response to cellular stress. The functional significance of ciliary resorption is highlighted in controlling the G1-S transition during cell cycle progression. Internal or external cues that trigger ciliary resorption initiate signaling cascades that regulate several downstream events including depolymerization of axonemal microtubules, dynamic changes in actin and the ciliary membrane, regulation of intraflagellar transport and posttranslational modifications of ciliary proteins. To ensure ciliary resorption, both the active disassembly of the cilium and the simultaneous inhibition of ciliary assembly must be coordinately regulated.
Collapse
Affiliation(s)
- Yinwen Liang
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Dan Meng
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bing Zhu
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Junmin Pan
- MOE Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China.
| |
Collapse
|
200
|
Rymut SM, Ivy T, Corey DA, Cotton CU, Burgess JD, Kelley TJ. Role of Exchange Protein Activated by cAMP 1 in Regulating Rates of Microtubule Formation in Cystic Fibrosis Epithelial Cells. Am J Respir Cell Mol Biol 2016; 53:853-62. [PMID: 25955407 DOI: 10.1165/rcmb.2014-0462oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The regulation of microtubule dynamics in cystic fibrosis (CF) epithelial cells and the consequences of reduced rates of microtubule polymerization on downstream CF cellular events, such as cholesterol accumulation, a marker of impaired intracellular transport, are explored here. It is identified that microtubules in both CF cell models and in primary CF nasal epithelial cells repolymerize at a slower rate compared with respective controls. Previous studies suggest a role for cAMP in modulating organelle transport in CF cells, implicating a role for exchange protein activated by cAMP (EPAC) 1, a regulator of microtubule elongation, as a potential mechanism. EPAC1 activity is reduced in CF cell models and in Cftr(-/-) mouse lung compared with respective non-CF controls. Stimulation of EPAC1 activity with the selective EPAC1 agonist, 8-cpt-2-O-Me-cAMP, stimulates microtubule repolymerization to wild-type rates in CF cells. EPAC1 activation also alleviates cholesterol accumulation in CF cells, suggesting a direct link between microtubule regulation and intracellular transport. To verify the relationship between transport and microtubule regulation, expression of the protein, tubulin polymerization-promoting protein, was knocked down in non-CF human tracheal (9/HTEo(-)) cells to mimic the microtubule dysregulation in CF cells. Transduced cells with short hairpin RNA targeting tubulin polymerization-promoting protein exhibit CF-like perinuclear cholesterol accumulation and other cellular manifestations of CF cells, thus supporting a role for microtubule regulation as a mechanism linking CFTR function to downstream cellular manifestation.
Collapse
Affiliation(s)
| | | | | | | | - James D Burgess
- 3 Chemistry, Case Western Reserve University, Cleveland, Ohio
| | - Thomas J Kelley
- Departments of 1 Pharmacology.,2 Pediatrics, and.,3 Chemistry, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|