1
|
Sakuragi S, Uchida T, Kato N, Zhao B, Takahashi T, Hattori A, Sakata Y, Soeda Y, Takashima A, Yoshimura H, Matsumoto G, Bannai H. Inducing aggresome and stable tau aggregation in Neuro2a cells with an optogenetic tool. Biophys Physicobiol 2024; 21:e210023. [PMID: 39963597 PMCID: PMC11832247 DOI: 10.2142/biophysico.bppb-v21.0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/26/2024] [Indexed: 02/20/2025] Open
Abstract
Tauopathy is a spectrum of diseases characterized by fibrillary tau aggregate formation in neurons and glial cells in the brain. Tau aggregation originates in the brainstem and entorhinal cortex and then spreads throughout the brain in Alzheimer's disease (AD), which is the most prevalent type of tauopathy. Understanding the mechanism by which locally developed tau pathology propagates throughout the brain is crucial for comprehending AD pathogenesis. Therefore, a novel model of tau pathology that artificially induces tau aggregation in targeted cells at specific times is essential. This study describes a novel optogenetic module, OptoTau, which is a human tau with the P301L mutation fused with a photosensitive protein CRY2olig, inducing various forms of tau according to the temporal pattern of blue light illumination pattern. Continuous blue light illumination for 12 h to Neuro2a cells that stably express OptoTau (OptoTauKI cells) formed clusters along microtubules, many of which eventually accumulated in aggresomes. Conversely, methanol-resistant tau aggregation was formed when alternating light exposure and darkness in 30-min cycles for 8 sets per day were repeated over 8 days. Methanol-resistant tau was induced more rapidly by repeating 5-min illumination followed by 25-min darkness over 24 h. These results indicate that OptoTau induced various tau aggregation stages based on the temporal pattern of blue light exposure. Thus, this technique exhibits potential as a novel approach to developing specific tau aggregation in targeted cells at desired time points.
Collapse
Affiliation(s)
- Shigeo Sakuragi
- Department of Electrical Engineering and Biosciences, School of Advanced Science and Engineering, Waseda University, Shinjuku-Ku, Tokyo 162-0056, Japan
| | - Tomoya Uchida
- Department of Electrical Engineering and Biosciences, School of Advanced Science and Engineering, Waseda University, Shinjuku-Ku, Tokyo 162-0056, Japan
| | - Naoki Kato
- Department of Electrical Engineering and Biosciences, School of Advanced Science and Engineering, Waseda University, Shinjuku-Ku, Tokyo 162-0056, Japan
| | - Boxiao Zhao
- Department of Electrical Engineering and Biosciences, School of Advanced Science and Engineering, Waseda University, Shinjuku-Ku, Tokyo 162-0056, Japan
| | - Toshiki Takahashi
- Department of Electrical Engineering and Biosciences, School of Advanced Science and Engineering, Waseda University, Shinjuku-Ku, Tokyo 162-0056, Japan
| | - Akito Hattori
- Department of Electrical Engineering and Biosciences, School of Advanced Science and Engineering, Waseda University, Shinjuku-Ku, Tokyo 162-0056, Japan
- Present address: Department of Medical Laboratory Science, Kitasato University School of Health Sciences, Minami-Uonuma, Niigata 949-7241, Japan
| | - Yoshihiro Sakata
- Department of Electrical Engineering and Biosciences, School of Advanced Science and Engineering, Waseda University, Shinjuku-Ku, Tokyo 162-0056, Japan
| | - Yoshiyuki Soeda
- Laboratory for Alzheimer’s Disease, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo 171-8588, Japan
| | - Akihiko Takashima
- Laboratory for Alzheimer’s Disease, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo 171-8588, Japan
| | - Hideaki Yoshimura
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Gen Matsumoto
- Department of Neurological Disease Control, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| | - Hiroko Bannai
- Department of Electrical Engineering and Biosciences, School of Advanced Science and Engineering, Waseda University, Shinjuku-Ku, Tokyo 162-0056, Japan
| |
Collapse
|
2
|
Soeda Y, Yoshimura H, Bannai H, Koike R, Shiiba I, Takashima A. Intracellular tau fragment droplets serve as seeds for tau fibrils. Structure 2024; 32:1793-1807.e6. [PMID: 39032487 DOI: 10.1016/j.str.2024.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 05/04/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024]
Abstract
Intracellular tau aggregation requires a local protein concentration increase, referred to as "droplets". However, the cellular mechanism for droplet formation is poorly understood. Here, we expressed OptoTau, a P301L mutant tau fused with CRY2olig, a light-sensitive protein that can form homo-oligomers. Under blue light exposure, OptoTau increased tau phosphorylation and was sequestered in aggresomes. Suppressing aggresome formation by nocodazole formed tau granular clusters in the cytoplasm. The granular clusters disappeared by discontinuing blue light exposure or 1,6-hexanediol treatment suggesting that intracellular tau droplet formation requires microtubule collapse. Expressing OptoTau-ΔN, a species of N-terminal cleaved tau observed in the Alzheimer's disease brain, formed 1,6-hexanediol and detergent-resistant tau clusters in the cytoplasm with blue light stimulation. These intracellular stable tau clusters acted as a seed for tau fibrils in vitro. These results suggest that tau droplet formation and N-terminal cleavage are necessary for neurofibrillary tangles formation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yoshiyuki Soeda
- Laboratory for Alzheimer's Disease, Department of Life Science, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo 171-8588, Japan.
| | - Hideaki Yoshimura
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroko Bannai
- School of Advanced Science and Engineering, Department of Electrical Engineering and Biosciences, Waseda University, 2-2 Wakamatsucho, Shinjuku-Ku, Tokyo 162-0056, Japan
| | - Riki Koike
- Laboratory for Alzheimer's Disease, Department of Life Science, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo 171-8588, Japan
| | - Isshin Shiiba
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo 171-8588, Japan
| | - Akihiko Takashima
- Laboratory for Alzheimer's Disease, Department of Life Science, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo 171-8588, Japan
| |
Collapse
|
3
|
Bahram Sangani N, Koetsier J, Mélius J, Kutmon M, Ehrhart F, Evelo CT, Curfs LMG, Reutelingsperger CP, Eijssen LMT. A novel insight into neurological disorders through HDAC6 protein-protein interactions. Sci Rep 2024; 14:14666. [PMID: 38918466 PMCID: PMC11199618 DOI: 10.1038/s41598-024-65094-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Due to its involvement in physiological and pathological processes, histone deacetylase 6 (HDAC6) is considered a promising pharmaceutical target for several neurological manifestations. However, the exact regulatory role of HDAC6 in the central nervous system (CNS) is still not fully understood. Hence, using a semi-automated literature screening technique, we systematically collected HDAC6-protein interactions that are experimentally validated and reported in the CNS. The resulting HDAC6 network encompassed 115 HDAC6-protein interactions divided over five subnetworks: (de)acetylation, phosphorylation, protein complexes, regulatory, and aggresome-autophagy subnetworks. In addition, 132 indirect interactions identified through HDAC6 inhibition were collected and categorized. Finally, to display the application of our HDAC6 network, we mapped transcriptomics data of Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis on the network and highlighted that in the case of Alzheimer's disease, alterations predominantly affect the HDAC6 phosphorylation subnetwork, whereas differential expression within the deacetylation subnetwork is observed across all three neurological disorders. In conclusion, the HDAC6 network created in the present study is a novel and valuable resource for the understanding of the HDAC6 regulatory mechanisms, thereby providing a framework for the integration and interpretation of omics data from neurological disorders and pharmacodynamic assessments.
Collapse
Affiliation(s)
- Nasim Bahram Sangani
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD, Maastricht, The Netherlands.
- GKC, Maastricht University Medical Centre, 6229 ER, Maastricht, The Netherlands.
| | - Jarno Koetsier
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
- GKC, Maastricht University Medical Centre, 6229 ER, Maastricht, The Netherlands
| | - Jonathan Mélius
- DataHub, Maastricht University & Maastricht UMC+, P. Debyelaan 15, 6229 HX, Maastricht, The Netherlands
| | - Martina Kutmon
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, 6200 MD, Maastricht, The Netherlands
| | - Friederike Ehrhart
- Department of Bioinformatics - BiGCaT, Research Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD, Maastricht, The Netherlands
| | - Chris T Evelo
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, 6200 MD, Maastricht, The Netherlands
- Department of Bioinformatics - BiGCaT, Research Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
| | - Leopold M G Curfs
- GKC, Maastricht University Medical Centre, 6229 ER, Maastricht, The Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
- GKC, Maastricht University Medical Centre, 6229 ER, Maastricht, The Netherlands
| | - Lars M T Eijssen
- Department of Bioinformatics - BiGCaT, Research Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
4
|
Körschgen H, Behl C. Aggresome-aggrephagy transition process: getting closer to the functional roles of HDAC6 and BAG3. Neural Regen Res 2024; 19:1181-1182. [PMID: 37905854 PMCID: PMC11467942 DOI: 10.4103/1673-5374.386407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/24/2023] [Accepted: 08/21/2023] [Indexed: 11/02/2023] Open
Affiliation(s)
- Hagen Körschgen
- The Autophagy Lab, Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christian Behl
- The Autophagy Lab, Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
5
|
Elabd S, Pauletto E, Solozobova V, Eickhoff N, Padrao N, Zwart W, Blattner C. TRIM25 targets p300 for degradation. Life Sci Alliance 2023; 6:e202301980. [PMID: 37770115 PMCID: PMC10539465 DOI: 10.26508/lsa.202301980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023] Open
Abstract
p300 is an important transcriptional co-factor. By stimulating the transfer of acetyl residues onto histones and several key transcription factors, p300 enhances transcriptional initiation and impacts cellular processes including cell proliferation and cell division. Despite its importance for cellular homeostasis, its regulation is poorly understood. We show that TRIM25, a member of the TRIM protein family, targets p300 for proteasomal degradation. However, despite TRIM25's RING domain and E3 activity, degradation of p300 by TRIM25 is independent of TRIM25-mediated p300 ubiquitination. Instead, TRIM25 promotes the interaction of p300 with dynein, which ensures a microtubule-dependent transport of p300 to cellular proteasomes. Through mediating p300 degradation, TRIM25 affects p300-dependent gene expression.
Collapse
Affiliation(s)
- Seham Elabd
- Institute for Biological and Chemical Systems - Biological Information Processing, Karlsruhe, Germany
- Human Physiology Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Eleonora Pauletto
- Institute for Biological and Chemical Systems - Biological Information Processing, Karlsruhe, Germany
| | - Valeria Solozobova
- Institute for Biological and Chemical Systems - Biological Information Processing, Karlsruhe, Germany
| | - Nils Eickhoff
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nuno Padrao
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Christine Blattner
- Institute for Biological and Chemical Systems - Biological Information Processing, Karlsruhe, Germany
| |
Collapse
|
6
|
Liao EE, Yang M, Nathan Kochen N, Vunnam N, Braun AR, Ferguson DM, Sachs JN. Proteasomal Stimulation by MK886 and Its Derivatives Can Rescue Tau-Induced Neurite Pathology. Mol Neurobiol 2023; 60:6133-6144. [PMID: 37428404 PMCID: PMC10529599 DOI: 10.1007/s12035-023-03417-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/31/2023] [Indexed: 07/11/2023]
Abstract
Proteasomal degradation of intrinsically disordered proteins, such as tau, is a critical component of proteostasis in both aging and neurodegenerative diseases. In this study, we investigated proteasomal activation by MK886 (MK). We previously identified MK as a lead compound capable of modulating tau oligomerization in a cellular FRET assay and rescuing P301L tau-induced cytotoxicity. We first confirmed robust proteasomal activation by MK using 20S proteasomal assays and a cellular proteasomal tau-GFP cleavage assay. We then show that MK treatment can significantly rescue tau-induced neurite pathology in differentiated SHSY5Y neurospheres. Due to this compelling result, we designed a series of seven MK analogs to determine if proteasomal activity is sensitive to structural permutations. Using the proteasome as the primary MOA, we examined tau aggregation, neurite outgrowth, inflammation, and autophagy assays to identify two essential substituents of MK that are required for compound activity: (1) removal of the N-chlorobenzyl group from MK negated both proteasomal and autophagic activity and reduced neurite outgrowth; and (2) removal of the indole-5-isopropyl group significantly improved neurite outgrowth and autophagy activity but reduced its anti-inflammatory capacity. Overall, our results suggest that the combination of proteasomal/autophagic stimulation and anti-inflammatory properties of MK and its derivatives can decrease tau-tau interactions and help rebalance dysfunctional proteostasis. Further development of MK to optimize its proteasomal, autophagic, and anti-inflammatory targets may lead to a novel therapeutic that would be beneficial in aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Elly E Liao
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Mu Yang
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Noah Nathan Kochen
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Nagamani Vunnam
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Anthony R Braun
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - David M Ferguson
- Dept. of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Jonathan N Sachs
- Dept. of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
7
|
Zhu Y, Feng M, Wang B, Zheng Y, Jiang D, Zhao L, Mamun MAA, Kang H, Nie H, Zhang X, Guo N, Qin S, Wang N, Liu H, Gao Y. New insights into the non-enzymatic function of HDAC6. Biomed Pharmacother 2023; 161:114438. [PMID: 37002569 DOI: 10.1016/j.biopha.2023.114438] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
Histone deacetylase 6 (HDAC6) is a class IIb histone deacetylase that contains two catalytic domains and a zinc-finger ubiquitin binding domain (ZnF-UBP) domain. The deacetylation function of HDAC6 has been extensively studied with common substrates such as α-tubulin, cortactin, and Hsp90. Apart from its deacetylase activity, HDAC6 ZnF-UBP binds to unanchored ubiquitin of specific sequences and serves as a carrier for transporting aggregated proteins. As a result, aggresomes are formed and protein degradation is facilitated by the autophagy-lysosome pathway. This HDAC6-dependent microtubule transport can be used by cells to assemble and activate inflammasomes, which play a critical role in immune regulation. Even viruses can benefit from the carrier of HDAC6 to assist in uncoating their surfaces during their infection cycle. However, HDAC6 is also capable of blocking virus invasion and replication in a non-enzymatic manner. Given these non-enzymatic functions, HDAC6 is closely associated with various diseases, including neurodegeneration, inflammasome-associated diseases, cancer, and viral infections. Small molecule inhibitors targeting the ubiquitin binding pocket of HDAC6 have been investigated. In this review, we focus on mechanisms in non-enzymatic functions of HDAC6 and discuss the rationality and prospects of therapeutic strategies by intervening the activation of HDAC6 ZnF-UBP in concrete diseases.
Collapse
Affiliation(s)
- Yuanzai Zhu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Mengkai Feng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Bo Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Yichao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Dandan Jiang
- Department of Pharmacy, People's Hospital of Henan Province, Zhengzhou University, Henan 450001, China
| | - Lijuan Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - M A A Mamun
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Huiqin Kang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Haiqian Nie
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Xiya Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ningjie Guo
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Shangshang Qin
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ning Wang
- The School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
8
|
Han SW, Ryu KY. Increased clearance of non-biodegradable polystyrene nanoplastics by exocytosis through inhibition of retrograde intracellular transport. JOURNAL OF HAZARDOUS MATERIALS 2022; 439:129576. [PMID: 35850071 DOI: 10.1016/j.jhazmat.2022.129576] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/28/2022] [Accepted: 07/09/2022] [Indexed: 06/15/2023]
Abstract
Nanoplastics (NPs) are derived from microplastics and may cause health problems. We previously showed that 100 nm polystyrene (PS)-NPs enter cells, including mouse embryonic fibroblasts (MEFs), and their intracellular accumulation induces inflammatory and oxidative stress. Moreover, PS-NP uptake was found to occur via endocytosis, and they accumulated mostly at the juxtanuclear position, but never within the nucleus. We speculated that PS-NPs were cleared from cells when they were no longer exposed to PS-NPs. However, the effects of PS-NPs on the cellular machinery remain unknown. The accumulation of PS-NPs at the juxtanuclear position may be due to retrograde transport along microtubules. To confirm this, we treated PS-NP-exposed MEFs with inhibitors of histone deacetylase 6 (HDAC6), dynein, or microtubule polymerization and found greatly diminished intracellular and juxtanuclear accumulation. Moreover, rapid clearance of PS-NPs was observed when MEFs were treated with an HDAC6 inhibitor. PS-NPs were removed by exocytosis, as confirmed by treatment with an exocytosis inhibitor. Furthermore, inhibiting the retrograde transport of PS-NPs alleviated the activation of the antioxidant response pathway, inflammatory and oxidative stress, and reactive oxygen species generation. In summary, inhibition of the retrograde transport of non-biodegradable PS-NPs leads to their rapid export by exocytosis, which may reduce their cytotoxicity.
Collapse
Affiliation(s)
- Seung-Woo Han
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Kwon-Yul Ryu
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea.
| |
Collapse
|
9
|
Zhang C, Huang C, Xia H, Xu H, Tang Q, Bi F. Autophagic sequestration of SQSTM1 disrupts the aggresome formation of ubiquitinated proteins during proteasome inhibition. Cell Death Dis 2022; 13:615. [PMID: 35840557 PMCID: PMC9287315 DOI: 10.1038/s41419-022-05061-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 01/21/2023]
Abstract
Aggresome formation is a protective cellular response to counteract proteasome dysfunction by sequestering misfolded proteins and reducing proteotoxic stress. Autophagic degradation of the protein aggregates is considered to be a key compensating mechanism for balancing proteostasis. However, the precise role of autophagy in proteasome inhibition-induced aggresome biogenesis remains unclear. Herein, we demonstrate that in the early stage of proteasome inhibition, the maturation of the autophagosome is suppressed, which facilitates aggresome formation of misfolded proteins. Proteasome inhibition-induced phosphorylation of SQSTM1 T269/S272 inhibits its autophagic receptor activity and promotes aggresome formation of misfolded proteins. Inhibiting SQSTM1 T269/S272 phosphorylation using Doramapimod aggravates proteasome inhibitor-mediated cell damage and tumor suppression. Taken together, our data reveal a negative effect of autophagy on aggresome biogenesis and cell damage upon proteasome inhibition. Our study suggests a novel therapeutic intervention for proteasome inhibitor-mediated tumor treatment.
Collapse
Affiliation(s)
- Chenliang Zhang
- grid.412901.f0000 0004 1770 1022Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Chen Huang
- grid.412901.f0000 0004 1770 1022Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Hongwei Xia
- grid.412901.f0000 0004 1770 1022Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Huanji Xu
- grid.412901.f0000 0004 1770 1022Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Qiulin Tang
- grid.412901.f0000 0004 1770 1022Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Feng Bi
- grid.412901.f0000 0004 1770 1022Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital of Sichuan University, Chengdu, China ,grid.412901.f0000 0004 1770 1022Department of Medical Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Guan X, Iyaswamy A, Sreenivasmurthy SG, Su C, Zhu Z, Liu J, Kan Y, Cheung KH, Lu J, Tan J, Li M. Mechanistic Insights into Selective Autophagy Subtypes in Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23073609. [PMID: 35408965 PMCID: PMC8998506 DOI: 10.3390/ijms23073609] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/01/2023] Open
Abstract
Eukaryotic cells possess a plethora of regulatory mechanisms to maintain homeostasis and ensure proper biochemical functionality. Autophagy, a central, conserved self-consuming process of the cell, ensures the timely degradation of damaged cellular components. Several studies have demonstrated the important roles of autophagy activation in mitigating neurodegenerative diseases, especially Alzheimer's disease (AD). However, surprisingly, activation of macroautophagy has not shown clinical efficacy. Hence, alternative strategies are urgently needed for AD therapy. In recent years, selective autophagy has been reported to be involved in AD pathology, and different subtypes have been identified, such as aggrephagy, mitophagy, reticulophagy, lipophagy, pexophagy, nucleophagy, lysophagy and ribophagy. By clarifying the underlying mechanisms governing these various subtypes, we may come to understand how to control autophagy to treat AD. In this review, we summarize the latest findings concerning the role of selective autophagy in the pathogenesis of AD. The evidence overwhelmingly suggests that selective autophagy is an active mechanism in AD pathology, and that regulating selective autophagy would be an effective strategy for controlling this pathogenesis.
Collapse
Affiliation(s)
- Xinjie Guan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (X.G.); (A.I.); (S.G.S.); (C.S.); (Z.Z.); (J.L.); (Y.K.); (K.-H.C.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Ashok Iyaswamy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (X.G.); (A.I.); (S.G.S.); (C.S.); (Z.Z.); (J.L.); (Y.K.); (K.-H.C.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Sravan Gopalkrishnashetty Sreenivasmurthy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (X.G.); (A.I.); (S.G.S.); (C.S.); (Z.Z.); (J.L.); (Y.K.); (K.-H.C.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Chengfu Su
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (X.G.); (A.I.); (S.G.S.); (C.S.); (Z.Z.); (J.L.); (Y.K.); (K.-H.C.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Zhou Zhu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (X.G.); (A.I.); (S.G.S.); (C.S.); (Z.Z.); (J.L.); (Y.K.); (K.-H.C.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Jia Liu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (X.G.); (A.I.); (S.G.S.); (C.S.); (Z.Z.); (J.L.); (Y.K.); (K.-H.C.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Yuxuan Kan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (X.G.); (A.I.); (S.G.S.); (C.S.); (Z.Z.); (J.L.); (Y.K.); (K.-H.C.)
| | - King-Ho Cheung
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (X.G.); (A.I.); (S.G.S.); (C.S.); (Z.Z.); (J.L.); (Y.K.); (K.-H.C.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Jiahong Lu
- State Key Lab of Quality Research in Chinese Medicine, University of Macau, Macao, China;
| | - Jieqiong Tan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410000, China
- Correspondence: (J.T.); (M.L.)
| | - Min Li
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (X.G.); (A.I.); (S.G.S.); (C.S.); (Z.Z.); (J.L.); (Y.K.); (K.-H.C.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
- Correspondence: (J.T.); (M.L.)
| |
Collapse
|
11
|
Du Y, Yang X, Li Z, Le W, Hao Y, Song Y, Wang F, Guan Y. HDAC6-mediated Hsp90 deacetylation reduces aggregation and toxicity of the protein alpha-synuclein by regulating chaperone-mediated autophagy. Neurochem Int 2021; 149:105141. [PMID: 34298079 DOI: 10.1016/j.neuint.2021.105141] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 06/26/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022]
Abstract
Histone deacetylase 6 (HDAC6) has been shown to control major cell response pathways to the cytotoxic ubiquitinated aggregates in some protein aggregation diseases. However, it is not well known whether HDAC6 affects the aggregation process of α-synuclein (α-syn) in Parkinson's disease (PD). Previously, we demonstrated that HDAC6 inhibition exacerbated the nigrostriatal dopamine neurodegeneration and up-regulated α-syn oligomers in a heat shock protein 90 (Hsp90)-dependent manner in PD mouse model. Here, we further showed that HDAC6 overexpression partly improved the behavior deficits of the PD model and alleviated the nigrostriatal dopamine (DA) neurons injury. Furthermore, HDAC6 was found to regulate α-syn oligomers levels through activation of chaperone-mediated autophagy (CMA). During this process, Hsp90 deacetylation mediated the crosstalk between HDAC6 and lysosome-associated membrane protein type 2A. Liquid chromatography-tandem mass spectrometry and mutational analysis showed that acetylation status Hsp90 at the K489 site was a strong determinant for HDAC6-induced CMA activation, α-syn oligomers levels, and cell survival in the cell model of PD. Therefore, our findings uncovered the mechanism of HDAC6 in the PD model that HDAC6 regulated α-syn oligomers levels and DA neurons survival partly through modulating CMA, and Hsp90 deacetylation at the K489 site mediated the crosstalk between HDAC6 and CMA. HDAC6 and its downstream effectors appear as key modulators of the cytotoxic α-syn aggregates, which deserve further investigations to evaluate their values as potential therapeutic targets in PD.
Collapse
Affiliation(s)
- Yunlan Du
- Department of Neurology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao Yang
- Department of Neurology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zezhi Li
- Department of Neurology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weidong Le
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial Hospital, Chengdu, China
| | - Yong Hao
- Department of Neurology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yeping Song
- Department of Neurology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fei Wang
- Department of Neurology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yangtai Guan
- Department of Neurology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Abstract
Neuroepigenetics, a new branch of epigenetics, plays an important role in the regulation of gene expression. Neuroepigenetics is associated with holistic neuronal function and helps in formation and maintenance of memory and learning processes. This includes neurodevelopment and neurodegenerative defects in which histone modification enzymes appear to play a crucial role. These modifications, carried out by acetyltransferases and deacetylases, regulate biologic and cellular processes such as apoptosis and autophagy, inflammatory response, mitochondrial dysfunction, cell-cycle progression and oxidative stress. Alterations in acetylation status of histone as well as non-histone substrates lead to transcriptional deregulation. Histone deacetylase decreases acetylation status and causes transcriptional repression of regulatory genes involved in neural plasticity, synaptogenesis, synaptic and neural plasticity, cognition and memory, and neural differentiation. Transcriptional deactivation in the brain results in development of neurodevelopmental and neurodegenerative disorders. Mounting evidence implicates histone deacetylase inhibitors as potential therapeutic targets to combat neurologic disorders. Recent studies have targeted naturally-occurring biomolecules and micro-RNAs to improve cognitive defects and memory. Multi-target drug ligands targeting HDAC have been developed and used in cell-culture and animal-models of neurologic disorders to ameliorate synaptic and cognitive dysfunction. Herein, we focus on the implications of histone deacetylase enzymes in neuropathology, their regulation of brain function and plausible involvement in the pathogenesis of neurologic defects.
Collapse
|
13
|
Wheeler JM, McMillan P, Strovas TJ, Liachko NF, Amlie-Wolf A, Kow RL, Klein RL, Szot P, Robinson L, Guthrie C, Saxton A, Kanaan NM, Raskind M, Peskind E, Trojanowski JQ, Lee VMY, Wang LS, Keene CD, Bird T, Schellenberg GD, Kraemer B. Activity of the poly(A) binding protein MSUT2 determines susceptibility to pathological tau in the mammalian brain. Sci Transl Med 2020; 11:11/523/eaao6545. [PMID: 31852801 DOI: 10.1126/scitranslmed.aao6545] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 06/20/2019] [Indexed: 12/15/2022]
Abstract
Brain lesions composed of pathological tau help to drive neurodegeneration in Alzheimer's disease (AD) and related tauopathies. Here, we identified the mammalian suppressor of tauopathy 2 (MSUT2) gene as a modifier of susceptibility to tau toxicity in two mouse models of tauopathy. Transgenic PS19 mice overexpressing tau, a model of AD, and lacking the Msut2 gene exhibited decreased learning and memory deficits, reduced neurodegeneration, and reduced accumulation of pathological tau compared to PS19 tau transgenic mice expressing Msut2 Conversely, Msut2 overexpression in 4RTauTg2652 tau transgenic mice increased pathological tau deposition and promoted the neuroinflammatory response to pathological tau. MSUT2 is a poly(A) RNA binding protein that antagonizes the canonical nuclear poly(A) binding protein PABPN1. In individuals with AD, MSUT2 abundance in postmortem brain tissue predicted an earlier age of disease onset. Postmortem AD brain tissue samples with normal amounts of MSUT2 showed elevated neuroinflammation associated with tau pathology. We observed co-depletion of MSUT2 and PABPN1 in postmortem brain samples from a subset of AD cases with higher tau burden and increased neuronal loss. This suggested that MSUT2 and PABPN1 may act together in a macromolecular complex bound to poly(A) RNA. Although MSUT2 and PABPN1 had opposing effects on both tau aggregation and poly(A) RNA tail length, we found that increased poly(A) tail length did not ameliorate tauopathy, implicating other functions of the MSUT2/PABPN1 complex in tau proteostasis. Our findings implicate poly(A) RNA binding proteins both as modulators of pathological tau toxicity in AD and as potential molecular targets for interventions to slow neurodegeneration in tauopathies.
Collapse
Affiliation(s)
- Jeanna M Wheeler
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Pamela McMillan
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Timothy J Strovas
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Nicole F Liachko
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Alexandre Amlie-Wolf
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca L Kow
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Ronald L Klein
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Patricia Szot
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA.,Mental Illness Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Linda Robinson
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Chris Guthrie
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Aleen Saxton
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Nicholas M Kanaan
- Department of Translational Sciences and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Murray Raskind
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA.,Mental Illness Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Elaine Peskind
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA.,Mental Illness Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M Y Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Thomas Bird
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.,Department of Neurology, University of Washington School of Medicine, Seattle, WA 98195, USA.,Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brian Kraemer
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA. .,Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA.,Department of Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
14
|
Lemos M, Stefanova N. Histone Deacetylase 6 and the Disease Mechanisms of α-Synucleinopathies. Front Synaptic Neurosci 2020; 12:586453. [PMID: 33041780 PMCID: PMC7518386 DOI: 10.3389/fnsyn.2020.586453] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/24/2020] [Indexed: 12/31/2022] Open
Abstract
The abnormal accumulation of α-Synuclein (α-Syn) is a prominent pathological feature in a group of diseases called α-Synucleinopathies, such as Parkinson’s disease, dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). The formation of Lewy bodies (LBs) and glial cytoplasmic inclusions (GCIs) in neurons and oligodendrocytes, respectively, is highly investigated. However, the molecular mechanisms behind α-Syn improper folding and aggregation remain unclear. Histone deacetylase 6 (HDAC6) is a Class II deacetylase, containing two active catalytic domains and a ubiquitin-binding domain. The properties of HDAC6 and its exclusive cytoplasmic localization allow HDAC6 to modulate the microtubule dynamics, acting as a specific α-tubulin deacetylase. Also, HDAC6 can bind ubiquitinated proteins, facilitating the formation of the aggresome, a cellular defense mechanism to cope with higher levels of misfolded proteins. Several studies report that the aggresome shares similarities in size and composition with LBs and GCIs. HDAC6 is found to co-localize with α-Syn in neurons and in oligodendrocytes, together with other aggresome-related proteins. The involvement of HDAC6 in several neurodegenerative diseases is already under discussion, however, the results obtained by modulating HDAC6 activity are not entirely conclusive. The main goal of this review is to summarize and critically discuss previous in vitro and in vivo data regarding the specific role of HDAC6 in the context of α-Syn accumulation and protein aggregation in α-Synucleinopathies.
Collapse
Affiliation(s)
- Miguel Lemos
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nadia Stefanova
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Wen X, An P, Li H, Zhou Z, Sun Y, Wang J, Ma L, Lu B. Tau Accumulation via Reduced Autophagy Mediates GGGGCC Repeat Expansion-Induced Neurodegeneration in Drosophila Model of ALS. Neurosci Bull 2020; 36:1414-1428. [PMID: 32500377 DOI: 10.1007/s12264-020-00518-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 03/21/2020] [Indexed: 12/21/2022] Open
Abstract
Expansions of trinucleotide or hexanucleotide repeats lead to several neurodegenerative disorders, including Huntington disease [caused by expanded CAG repeats (CAGr) in the HTT gene], and amyotrophic lateral sclerosis [ALS, possibly caused by expanded GGGGCC repeats (G4C2r) in the C9ORF72 gene], of which the molecular mechanisms remain unclear. Here, we demonstrated that lowering the Drosophila homologue of tau protein (dtau) significantly rescued in vivo neurodegeneration, motor performance impairments, and the shortened life-span in Drosophila expressing expanded CAGr or expanded G4C2r. Expression of human tau (htau4R) restored the disease-related phenotypes that had been mitigated by the loss of dtau, suggesting an evolutionarily-conserved role of tau in neurodegeneration. We further revealed that G4C2r expression increased tau accumulation by inhibiting autophagosome-lysosome fusion, possibly due to lowering the level of BAG3, a regulator of autophagy and tau. Taken together, our results reveal a novel mechanism by which expanded G4C2r causes neurodegeneration via an evolutionarily-conserved mechanism. Our findings provide novel autophagy-related mechanistic insights into C9ORF72-ALS and possible entry points to disease treatment.
Collapse
Affiliation(s)
- Xue Wen
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Ping An
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Hexuan Li
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Zijian Zhou
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yimin Sun
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jian Wang
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Lixiang Ma
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Boxun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
16
|
Singh JK, Hutt DM, Tait B, Guy NC, Sivils JC, Ortiz NR, Payan AN, Komaragiri SK, Owens JJ, Culbertson D, Blair LJ, Dickey C, Kuo SY, Finley D, Dyson HJ, Cox MB, Chaudhary J, Gestwicki JE, Balch WE. Management of Hsp90-Dependent Protein Folding by Small Molecules Targeting the Aha1 Co-Chaperone. Cell Chem Biol 2020; 27:292-305.e6. [PMID: 32017918 PMCID: PMC7144688 DOI: 10.1016/j.chembiol.2020.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/18/2019] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Hsp90 plays an important role in health and is a therapeutic target for managing misfolding disease. Compounds that disrupt co-chaperone delivery of clients to Hsp90 target a subset of Hsp90 activities, thereby minimizing the toxicity of pan-Hsp90 inhibitors. Here, we have identified SEW04784 as a first-in-class inhibitor of the Aha1-stimulated Hsp90 ATPase activity without inhibiting basal Hsp90 ATPase. Nuclear magnetic resonance analysis reveals that SEW84 binds to the C-terminal domain of Aha1 to weaken its asymmetric binding to Hsp90. Consistent with this observation, SEW84 blocks Aha1-dependent Hsp90 chaperoning activities, including the in vitro and in vivo refolding of firefly luciferase, and the transcriptional activity of the androgen receptor in cell-based models of prostate cancer and promotes the clearance of phosphorylated tau in cellular and tissue models of neurodegenerative tauopathy. We propose that SEW84 provides a novel lead scaffold for developing therapeutic approaches to treat proteostatic disease.
Collapse
Affiliation(s)
- Jay K Singh
- Department of Molecular Medicine, Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Darren M Hutt
- Department of Molecular Medicine, Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bradley Tait
- Brad Tait Enterprise LLC, 80 Christian Way, North Andover, MA 01845, USA
| | - Naihsuan C Guy
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Jeffrey C Sivils
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Nina R Ortiz
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Ashley N Payan
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79902, USA
| | | | | | - David Culbertson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Laura J Blair
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, University of South Florida, Tampa, FL 33613, USA
| | - Chad Dickey
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, University of South Florida, Tampa, FL 33613, USA
| | - Szu Yu Kuo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Dan Finley
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - H Jane Dyson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Marc B Cox
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Jaideep Chaudhary
- School of Arts and Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - William E Balch
- Department of Molecular Medicine, Skaggs Institute of Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
17
|
Targeting Aggrephagy for the Treatment of Alzheimer's Disease. Cells 2020; 9:cells9020311. [PMID: 32012902 PMCID: PMC7072705 DOI: 10.3390/cells9020311] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/25/2020] [Accepted: 01/26/2020] [Indexed: 12/17/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common neurodegenerative diseases in older individuals with specific neuropsychiatric symptoms. It is a proteinopathy, pathologically characterized by the presence of misfolded protein (Aβ and Tau) aggregates in the brain, causing progressive dementia. Increasing studies have provided evidence that the defect in protein-degrading systems, especially the autophagy-lysosome pathway (ALP), plays an important role in the pathogenesis of AD. Recent studies have demonstrated that AD-associated protein aggregates can be selectively recognized by some receptors and then be degraded by ALP, a process termed aggrephagy. In this study, we reviewed the role of aggrephagy in AD development and discussed the strategy of promoting aggrephagy using small molecules for the treatment of AD.
Collapse
|
18
|
Pharmacological intervention of histone deacetylase enzymes in the neurodegenerative disorders. Life Sci 2020; 243:117278. [PMID: 31926248 DOI: 10.1016/j.lfs.2020.117278] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023]
Abstract
Reversal of aging symptoms and related disorders are the challenging task where epigenetic is a crucial player that includes DNA methylation, histone modification; chromatin remodeling and regulation that are linked to the progression of various neurodegenerative disorders (NDDs). Overexpression of various histone deacetylase (HDACs) can activate Glycogen synthase kinase 3 which promotes the hyperphosphorylation of tau and inhibits its degradation. While HDAC is important for maintaining the neuronal morphology and brain homeostasis, at the same time, these enzymes are promoting neurodegeneration, if it is deregulated. Different experimental models have also confirmed the neuroprotective effects caused by HDAC enzymes through the regulation of neuronal apoptosis, inflammatory response, DNA damage, cell cycle regulation, and metabolic dysfunction. Apart from transcriptional regulation, protein-protein interaction, histone post-translational modifications, deacetylation mechanism of non-histone protein and direct association with disease proteins have been linked to neuronal imbalance. Histone deacetylases inhibitors (HDACi) can be able to alter gene expression and shown its efficacy on experimental models, and in clinical trials for NDD's and found to be a very promising therapeutic agent with certain limitation, for instance, non-specific target effect, isoform-selectivity, specificity, and limited number of predicted biomarkers. Herein, we discussed (i) the catalytic mechanism of the deacetylation process of various HDAC's in in vivo and in vitro experimental models, (ii) how HDACs are participating in neuroprotection as well as in neurodegeneration, (iii) a comprehensive role of HDACi in maintaining neuronal homeostasis and (iv) therapeutic role of biomolecules to modulate HDACs.
Collapse
|
19
|
Piatnitskaia S, Takahashi M, Kitaura H, Katsuragi Y, Kakihana T, Zhang L, Kakita A, Iwakura Y, Nawa H, Miura T, Ikeuchi T, Hara T, Fujii M. USP10 is a critical factor for Tau-positive stress granule formation in neuronal cells. Sci Rep 2019; 9:10591. [PMID: 31332267 PMCID: PMC6646309 DOI: 10.1038/s41598-019-47033-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 07/08/2019] [Indexed: 12/31/2022] Open
Abstract
Tau aggregates in neurons of brain lesions is a hallmark pathology of tauopathies, including Alzheimer’s disease (AD). Recent studies suggest that the RNA-binding protein TIA1 initiates Tau aggregation by inducing the formation of stress granules (SGs) containing Tau. SGs are stress-inducible cytoplasmic protein aggregates containing many RNA-binding proteins that has been implicated as an initial site of multiple pathogenic protein aggregates in several neurodegenerative diseases. In this study, we found that ubiquitin-specific protease 10 (USP10) is a critical factor for the formation of Tau/TIA1/USP10-positive SGs. Proteasome inhibition or TIA1-overexpression in HT22 neuronal cells induced the formation of TIA1/Tau-positive SGs, and the formations were severely attenuated by depletion of USP10. In addition, the overexpression of USP10 without stress stimuli in HT22 cells induced TIA1/Tau/USP10-positive SGs in a deubiquitinase-independent manner. In AD brain lesions, USP10 was colocalized with Tau aggregates in the cell body of neurons. The present findings suggest that USP10 plays a key role in the initiation of pathogenic Tau aggregation in AD through SG formation.
Collapse
Affiliation(s)
- Svetlana Piatnitskaia
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Masahiko Takahashi
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Hiroki Kitaura
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Yoshinori Katsuragi
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Taichi Kakihana
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Lu Zhang
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Yuriko Iwakura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Takeshi Miura
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Toshifumi Hara
- Department of Medicinal Biochemistry, School of Pharmacy, Aichi Gakuin University, Nagoya, Japan
| | - Masahiro Fujii
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan.
| |
Collapse
|
20
|
Tau Protein Squired by Molecular Chaperones During Alzheimer’s Disease. J Mol Neurosci 2018; 66:356-368. [DOI: 10.1007/s12031-018-1174-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 09/14/2018] [Indexed: 01/19/2023]
|
21
|
Thomas EA, D'Mello SR. Complex neuroprotective and neurotoxic effects of histone deacetylases. J Neurochem 2018; 145:96-110. [PMID: 29355955 DOI: 10.1111/jnc.14309] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/05/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
By their ability to shatter quality of life for both patients and caregivers, neurodegenerative diseases are the most devastating of human disorders. Unfortunately, there are no effective or long-terms treatments capable of slowing down the relentless loss of neurons in any of these diseases. One impediment is the lack of detailed knowledge of the molecular mechanisms underlying the processes of neurodegeneration. While some neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, are mostly sporadic in nature, driven by both environment and genetic susceptibility, many others, including Huntington's disease, spinocerebellar ataxias, and spinal-bulbar muscular atrophy, are genetically inherited disorders. Surprisingly, given their different roots and etiologies, both sporadic and genetic neurodegenerative disorders have been linked to disease mechanisms involving histone deacetylase (HDAC) proteins, which consists of 18 family members with diverse functions. While most studies have implicated certain HDAC subtypes in promoting neurodegeneration, a substantial body of literature suggests that other HDAC proteins can preserve neuronal viability. Of particular interest, however, is the recent realization that a single HDAC subtype can have both neuroprotective and neurotoxic effects. Diverse mechanisms, beyond transcriptional regulation have been linked to these effects, including deacetylation of non-histone proteins, protein-protein interactions, post-translational modifications of the HDAC proteins themselves and direct interactions with disease proteins. The roles of these HDACs in both sporadic and genetic neurodegenerative diseases will be discussed in the current review.
Collapse
Affiliation(s)
- Elizabeth A Thomas
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | - Santosh R D'Mello
- Department of Biological Sciences, Southern Methodist University, Dallas, Texas, USA
| |
Collapse
|
22
|
Khurana N, Kim H, Chandra PK, Talwar S, Sharma P, Abdel-Mageed AB, Sikka SC, Mondal D. Multimodal actions of the phytochemical sulforaphane suppress both AR and AR-V7 in 22Rv1 cells: Advocating a potent pharmaceutical combination against castration-resistant prostate cancer. Oncol Rep 2017; 38:2774-2786. [PMID: 28901514 PMCID: PMC5780030 DOI: 10.3892/or.2017.5932] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/27/2017] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) cells expressing full-length androgen receptor (AR-FL) are susceptible to androgen deprivation therapy (ADT). However, outgrowth of castration-resistant prostate cancer (CRPC) can occur due to the expression of constitutively active (ligand-independent) AR splice variants, particularly AR-V7. We previously demonstrated that sulforaphane (SFN), an isothiocyanate phytochemical, can decrease AR-FL levels in the PCa cell lines, LNCaP and C4-2B. Here, we examined the efficacy of SFN in targeting both AR-FL and AR-V7 in the CRPC cell line, CWR22Rv1 (22Rv1). MTT cell viability, wound-heal assay, and colony forming unit (CFU) measurements revealed that 22Rv1 cells are resistant to the anti-androgen, enzalutamide (ENZ). However, co-exposure to SFN sensitized these cells to the potent anticancer effects of ENZ (P<0.05). Immunoblot analyses showed that SFN (5–20 µM) rapidly decreases both AR-FL and AR-V7 levels, and immunofluorescence microscopy (IFM) depicted decreased AR in both cytoplasm and nucleus with SFN treatment. SFN increased both ubiquitination and proteasomal activity in 22Rv1 cells. Studies using a protein synthesis inhibitor (cycloheximide) or a proteasomal inhibitor (MG132) indicated that SFN increases both ubiquitin-mediated aggregation and subsequent proteasomal-degradation of AR proteins. Previous studies reported that SFN inhibits the chaperone activity of heat-shock protein 90 (Hsp90) and induces the nuclear factor erythroid-2-like 2 (Nrf2) transcription factor. Therefore, we investigated whether the Hsp90 inhibitor, ganetespib (G) or the Nrf2 activator, bardoxolone methyl (BM) can similarly suppress AR levels in 22Rv1 cells. Low doses of G and BM, alone or in combination, decreased both AR-FL and AR-V7 levels, and combined exposure to G+BM sensitized 22Rv1 cells to ENZ. Therefore, adjunct treatment with the phytochemical SFN or a safe pharmaceutical combination of G+BM may be effective against CRPC cells, especially those expressing AR-V7.
Collapse
Affiliation(s)
- Namrata Khurana
- Department of Urology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hogyoung Kim
- Department of Urology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Partha K Chandra
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Sudha Talwar
- Department of Urology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Pankaj Sharma
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Asim B Abdel-Mageed
- Department of Urology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Suresh C Sikka
- Department of Urology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Debasis Mondal
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
23
|
UCH-L1 Inhibition Suppresses tau Aggresome Formation during Proteasomal Impairment. Mol Neurobiol 2017; 55:3812-3821. [PMID: 28540657 DOI: 10.1007/s12035-017-0558-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
Abstract
In conditions of proteasomal impairment, the damaged or misfolded proteins, collectively known as aggresome, can accumulate in the perinuclear space and be subsequently eliminated by autophagy. Abnormal aggregation of microtubule-associated protein tau in the cytoplasm is a common neuropathological feature of tauopathies. The deficiency in ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), a proteasomal deubiquitinating enzyme, is closely related to tau aggregation; however, the associated mechanisms remain unclear. Here, we showed that UCH-L1 inhibition interrupts proteasomal impairment-induced tau aggresome formation. By reducing the production of lysine (K63)-linked ubiquitin chains, UCH-L1 inhibition decreases HDAC6 deacetylase activity and attenuates the interaction of HDAC6 and tau protein, finally leading to tau aggresome formation impairment. All these results indicated that UCH-L1 plays a key role in the process of tau aggresome formation by regulating HDAC6 deacetylase activity and implied that UCH-L1 may act as a signaling molecule to coordinate the effects of the ubiquitin-proteasome system and the autophagy-lysosome pathway, which mediate protein aggregates degradation in the cytoplasm.
Collapse
|
24
|
Li ZY, Zhang C, Zhang Y, Chen L, Chen BD, Li QZ, Zhang XJ, Li WP. A novel HDAC6 inhibitor Tubastatin A: Controls HDAC6-p97/VCP-mediated ubiquitination-autophagy turnover and reverses Temozolomide-induced ER stress-tolerance in GBM cells. Cancer Lett 2017; 391:89-99. [PMID: 28131906 DOI: 10.1016/j.canlet.2017.01.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 01/21/2023]
Abstract
Temozolomide (TMZ) is the cornerstone of therapy for glioblastoma multiforme (GBM). However, its efficacy is limited due to the development of multidrug resistance (MDR). In this study, we first identified the occurrence of ER stress-tolerance (ERST) in glioma cells and confirmed that ERST was positively correlated with TMZ resistance. We further showed that the seesaw-effect of HDAC6-p97/VCP (increased HDAC6 and decreased p97/VCP) in glioma cells was crucial to ERST-associated TMZ resistance. Moreover, the combination treatment of Tubastatin A (TUB, a selective inhibitor of HDAC6) and TMZ synergistically overcame ERST, reduced cell viability and induced apoptosis in TMZ-resistant glioma cells. TUB and TMZ triggered pro-apoptotic signals of the unfolded protein response (UPR) and ER stress and reversed the ratio between HDAC6 and p97/VCP, which potentially attenuated the activation of heat shock proteins and mediated the reversal of ERST. The combination treatment also triggered the dissociation of Dynein-HDAC6 and attenuation of the Dynein-Dynactin motor complex. In addition, this treatment induced HDAC6-p97/VCP-mediated ubiquitination-autophagy turnover, which was involved in the degradation and clearance of ubiquitinated misfolded proteins. This effect could be partially reversed by HDAC6 KO and/or p97/VCP overexpression. Therefore, we proposed that glioma cells optimized the clearance of ubiquitinated misfolded proteins via the reinforcement of HDAC6-facilitated autophagy and attenuation of the p97/VCP-mediated ubiquitin-proteasome system (UPS). In conclusion, our findings showed that the balance of HDAC6-p97/VCP was crucial to ERST-associated TMZ resistance and that HDAC6 inhibition might be a synergistic target and strategy along with TMZ for the improvement of clinical glioma treatment.
Collapse
Affiliation(s)
- Zong-Yang Li
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Ce Zhang
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Yuan Zhang
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Lei Chen
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Bao-Dong Chen
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Qing-Zhong Li
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Xie-Jun Zhang
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China
| | - Wei-Ping Li
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002# Sungang Road, Futian District, Shenzhen 518035, China.
| |
Collapse
|
25
|
Hyttinen JM, Amadio M, Viiri J, Pascale A, Salminen A, Kaarniranta K. Clearance of misfolded and aggregated proteins by aggrephagy and implications for aggregation diseases. Ageing Res Rev 2014; 18:16-28. [PMID: 25062811 DOI: 10.1016/j.arr.2014.07.002] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/02/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022]
Abstract
Processing of misfolded proteins is important in order for the cell to maintain its normal functioning and homeostasis. Three systems control the quality of proteins: chaperone-mediated refolding, proteasomal degradation of ubiquitinated proteins, and finally, when the two others fail, aggrephagy, as selective form of autophagy, degrades ubiquitin-labelled aggregated cargos. In this route misfolded proteins gradually form larger aggregates, aggresomes and they eventually become double membrane-wrapped organelles called autophagosomes, which become degraded when they fuse to lysosomes, for reuse by the cell. The stages, the main molecules participating in the process, and the regulation of aggrephagy are discussed here, as is the role of protein aggregation in protein accumulation diseases. In particular, we emphasize that both Alzheimer's disease and age-related macular degeneration, two of the most common pathologies in the aged, are characterized by altered protein clearance and deposits. Based on the hypothesis that manipulations of autophagy may be potentially useful in these and other aggregation-related diseases, we will discuss some promising therapeutic strategies to counteract protein aggregates-induced cellular toxicity.
Collapse
|
26
|
Calderilla-Barbosa L, Seibenhener ML, Du Y, Diaz-Meco MT, Moscat J, Yan J, Wooten MW, Wooten MC. Interaction of SQSTM1 with the motor protein dynein--SQSTM1 is required for normal dynein function and trafficking. J Cell Sci 2014; 127:4052-63. [PMID: 25015291 DOI: 10.1242/jcs.152363] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The dynein motor protein complex is required for retrograde transport of vesicular cargo and for transport of aggregated proteins along microtubules for processing and degradation at perinuclear aggresomes. Disruption of this process leads to dysfunctional endosome accumulation and increased protein aggregation in the cell cytoplasm, both pathological features of neurodegenerative diseases. However, the exact mechanism of dynein functionality in these pathways is still being elucidated. Here, we show that the scaffolding protein SQSTM1 directly interacts with dynein through a previously unidentified dynein-binding site. This interaction is independent of HDAC6, a known interacting protein of both SQSTM1 and dynein. However, knockdown of HDAC6 increases the interaction of SQSTM1 with dynein, indicating a possible competitive interaction. Using different dynein cargoes, we show that SQSTM1 is required for proper dynein motility and trafficking along microtubules. Based on our results, we propose a new model of competitive interaction between SQSTM1 and HDAC6 with dynein. In this model, SQSTM1 would not only affect the association of polyubiquitylated protein aggregates and endosomes with dynein, but would also be required for normal dynein function.
Collapse
Affiliation(s)
- Luis Calderilla-Barbosa
- Department of Biological Sciences, 331 Funchess Hall, Auburn University, Auburn, AL 36849, USA
| | - M Lamar Seibenhener
- Department of Biological Sciences, 331 Funchess Hall, Auburn University, Auburn, AL 36849, USA
| | - Yifeng Du
- Department of Biological Sciences, 331 Funchess Hall, Auburn University, Auburn, AL 36849, USA
| | - Maria-Theresa Diaz-Meco
- 10901 North Torrey Pines Road, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Jorge Moscat
- 10901 North Torrey Pines Road, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Jin Yan
- Graduate Center for Toxicology, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Marie W Wooten
- Department of Biological Sciences, 331 Funchess Hall, Auburn University, Auburn, AL 36849, USA
| | - Michael C Wooten
- Department of Biological Sciences, 331 Funchess Hall, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
27
|
Yan J. Interplay between HDAC6 and its interacting partners: essential roles in the aggresome-autophagy pathway and neurodegenerative diseases. DNA Cell Biol 2014; 33:567-80. [PMID: 24932665 DOI: 10.1089/dna.2013.2300] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cytoplasmic localization and possession of two deacetylase domains and a ubiquitin-binding domain make histone deacetylase 6 (HDAC6) a unique histone deacetylase. HDAC6 interacts with a number of proteins in the cytoplasm. Some of these proteins can be deacetylated by HDAC6 deacetylase activity. Others can affect HDAC6 functions by modulating its catalytic activity or ubiquitin-binding capability. Over the last decade, HDAC6 has been shown to play important roles in the aggresome-autophagy pathway, which selectively targets on protein aggregates or damaged organelles for their accumulation and clearance in cells. HDAC6-interacting partners are integral components in this pathway with regard to their regulatory roles through interaction with HDAC6. The aggresome-autophagy pathway appears to be an attractive therapeutic target for the treatment of neurodegenerative diseases as accumulation of protein aggregates are hallmarks in these diseases. In the current review, I discuss the molecular details of how HDAC6 and its interacting partners regulate each individual step in the aggresome-autophagy pathway and also provide perspectives of how HDAC6 can be targeted in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Jin Yan
- Department of Biological Sciences, Auburn University , Auburn, Alabama
| |
Collapse
|
28
|
Salemi LM, Almawi AW, Lefebvre KJ, Schild-Poulter C. Aggresome formation is regulated by RanBPM through an interaction with HDAC6. Biol Open 2014; 3:418-30. [PMID: 24795145 PMCID: PMC4058076 DOI: 10.1242/bio.20147021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In conditions of proteasomal impairment, the build-up of damaged or misfolded proteins activates a cellular response leading to the recruitment of damaged proteins into perinuclear aggregates called aggresomes. Aggresome formation involves the retrograde transport of cargo proteins along the microtubule network and is dependent on the histone deacetylase HDAC6. Here we show that ionizing radiation (IR) promotes Ran-Binding Protein M (RanBPM) relocalization into discrete perinuclear foci where it co-localizes with aggresome components ubiquitin, dynein and HDAC6, suggesting that the RanBPM perinuclear clusters correspond to aggresomes. RanBPM was also recruited to aggresomes following treatment with the proteasome inhibitor MG132 and the DNA-damaging agent etoposide. Strikingly, aggresome formation by HDAC6 was markedly impaired in RanBPM shRNA cells, but was restored by re-expression of RanBPM. RanBPM was found to interact with HDAC6 and to inhibit its deacetylase activity. This interaction was abrogated by a RanBPM deletion of its LisH/CTLH domain, which also prevented aggresome formation, suggesting that RanBPM promotes aggresome formation through an association with HDAC6. Our results suggest that RanBPM regulates HDAC6 activity and is a central regulator of aggresome formation.
Collapse
Affiliation(s)
- Louisa M Salemi
- Robarts Research Institute, The University of Western Ontario, London, ON N6A 5B7, Canada Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada
| | - Ahmad W Almawi
- Robarts Research Institute, The University of Western Ontario, London, ON N6A 5B7, Canada Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada
| | - Karen J Lefebvre
- Robarts Research Institute, The University of Western Ontario, London, ON N6A 5B7, Canada Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada
| | - Caroline Schild-Poulter
- Robarts Research Institute, The University of Western Ontario, London, ON N6A 5B7, Canada Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
29
|
Gal J, Chen J, Barnett KR, Yang L, Brumley E, Zhu H. HDAC6 regulates mutant SOD1 aggregation through two SMIR motifs and tubulin acetylation. J Biol Chem 2013; 288:15035-45. [PMID: 23580651 DOI: 10.1074/jbc.m112.431957] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) is a tubulin deacetylase that regulates protein aggregation and turnover. Mutations in Cu/Zn superoxide dismutase (SOD1) linked to familial amyotrophic lateral sclerosis (ALS) make the mutant protein prone to aggregation. However, the role of HDAC6 in mutant SOD1 aggregation and the ALS etiology is unclear. Here we report that HDAC6 knockdown increased mutant SOD1 aggregation in cultured cells. Different from its known role in mediating the degradation of poly-ubiquitinated proteins, HDAC6 selectively interacted with mutant SOD1 via two motifs similar to the SOD1 mutant interaction region (SMIR) that we identified previously in p62/sequestosome 1. Expression of the aggregation-prone mutant SOD1 increased α-tubulin acetylation, and the acetylation-mimicking K40Q α-tubulin mutant promoted mutant SOD1 aggregation. Our results suggest that ALS-linked mutant SOD1 can modulate HDAC6 activity and increase tubulin acetylation, which, in turn, facilitates the microtubule- and retrograde transport-dependent mutant SOD1 aggregation. HDAC6 impairment might be a common feature in various subtypes of ALS.
Collapse
Affiliation(s)
- Jozsef Gal
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | |
Collapse
|
30
|
Liu Y, Su Y, Wang J, Sun S, Wang T, Qiao X, Run X, Li H, Liang Z. Rapamycin decreases tau phosphorylation at Ser214 through regulation of cAMP-dependent kinase. Neurochem Int 2013; 62:458-67. [DOI: 10.1016/j.neuint.2013.01.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 12/26/2012] [Accepted: 01/09/2013] [Indexed: 01/25/2023]
|
31
|
Dopamine D2 receptor antagonism suppresses tau aggregation and neurotoxicity. Biol Psychiatry 2013; 73:464-71. [PMID: 23140663 PMCID: PMC3570611 DOI: 10.1016/j.biopsych.2012.08.027] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 08/29/2012] [Accepted: 08/30/2012] [Indexed: 01/11/2023]
Abstract
BACKGROUND Tauopathies, including Alzheimer's disease and frontotemporal dementia, are diseases characterized by the formation of pathological tau protein aggregates in the brain and progressive neurodegeneration. Presently no effective disease-modifying treatments exist for tauopathies. METHODS To identify drugs targeting tau neurotoxicity, we have used a Caenorhabditis elegans model of tauopathy to screen a drug library containing 1120 compounds approved for human use for the ability to suppress tau-induced behavioral effects. RESULTS One compound, the typical antipsychotic azaperone, improved the motility of tau transgenic worms, reduced levels of insoluble tau, and was protective against neurodegeneration. We found that azaperone reduces insoluble tau in a human cell culture model of tau aggregation and that other antipsychotic drugs (flupenthixol, perphenazine, and zotepine) also ameliorate the effects of tau expression in both models. CONCLUSIONS Reduction of dopamine signaling through the dopamine D2 receptor with the use of gene knockouts in Caenorhabditis elegans or RNA interference knockdown in human cell culture has similar protective effects against tau toxicity. These results suggest dopamine D2 receptor antagonism holds promise as a potential neuroprotective strategy for targeting tau aggregation and neurotoxicity.
Collapse
|
32
|
Hebron ML, Algarzae NK, Lonskaya I, Moussa C. Fractalkine signaling and Tau hyper-phosphorylation are associated with autophagic alterations in lentiviral Tau and Aβ1-42 gene transfer models. Exp Neurol 2013; 251:127-38. [PMID: 23333589 DOI: 10.1016/j.expneurol.2013.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 01/03/2013] [Accepted: 01/09/2013] [Indexed: 12/17/2022]
Abstract
Tau hyper-phosphorylation (p-Tau) and neuro-inflammation are hallmarks of neurodegeneration. Previous findings suggest that microglial activation via CX3CL1 promotes p-Tau. We examined inflammation and autophagic p-Tau clearance in lentiviral Tau and mutant P301L expressing rats and used lentiviral Aβ1-42 to induce p-Tau. Lentiviral Tau or P301L expression significantly increased caspase-3 activity and TNF-α, but CX3CL1 was significantly higher in animals expressing Tau compared to P301L. Lentiviral Aβ1-42 induced p-Tau 4 weeks post-injection, and increased caspase-3 activation (8-fold) and TNF-α levels. Increased levels of ADAM-10/17 were also detected with p-Tau. IL-6 levels were increased but CX3CL1 did not change in the absence of p-Tau (2 weeks); however, p-Tau reversed these effects, which were associated with increased microglial activity. We observed changes in autophagic markers, including accumulation of autophagic vacuoles (AVs) and p-Tau accumulation in autophagosomes but not lysosomes, suggesting alteration of autophagy. Taken together, microglial activation may promote p-Tau independent of total Tau levels via CX3CL1 signaling, which seems to depend on interaction with inflammatory markers, mainly IL-6. The simultaneous change in autophagy and CX3CL1 signaling suggests communication between microglia and neurons, raising the possibility that accumulation of intraneuronal amyloid, due to lack of autophagic clearance, may lead microglia activation to promote p-Tau as a tag for phagocytic degradation.
Collapse
Affiliation(s)
- Michaeline L Hebron
- Department of Neuroscience, Georgetown University Medical Center, Washington D.C. 20007, USA; Department of Biochemistry and Cell Biology, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - Norah K Algarzae
- Department of Neuroscience, Georgetown University Medical Center, Washington D.C. 20007, USA; Department of Biochemistry and Cell Biology, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - Irina Lonskaya
- Department of Neuroscience, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - Charbel Moussa
- Department of Neuroscience, Georgetown University Medical Center, Washington D.C. 20007, USA.
| |
Collapse
|
33
|
Chondrogianni N, Petropoulos I, Grimm S, Georgila K, Catalgol B, Friguet B, Grune T, Gonos ES. Protein damage, repair and proteolysis. Mol Aspects Med 2012; 35:1-71. [PMID: 23107776 DOI: 10.1016/j.mam.2012.09.001] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 09/26/2012] [Indexed: 01/10/2023]
Abstract
Proteins are continuously affected by various intrinsic and extrinsic factors. Damaged proteins influence several intracellular pathways and result in different disorders and diseases. Aggregation of damaged proteins depends on the balance between their generation and their reversal or elimination by protein repair systems and degradation, respectively. With regard to protein repair, only few repair mechanisms have been evidenced including the reduction of methionine sulfoxide residues by the methionine sulfoxide reductases, the conversion of isoaspartyl residues to L-aspartate by L-isoaspartate methyl transferase and deglycation by phosphorylation of protein-bound fructosamine by fructosamine-3-kinase. Protein degradation is orchestrated by two major proteolytic systems, namely the lysosome and the proteasome. Alteration of the function for both systems has been involved in all aspects of cellular metabolic networks linked to either normal or pathological processes. Given the importance of protein repair and degradation, great effort has recently been made regarding the modulation of these systems in various physiological conditions such as aging, as well as in diseases. Genetic modulation has produced promising results in the area of protein repair enzymes but there are not yet any identified potent inhibitors, and, to our knowledge, only one activating compound has been reported so far. In contrast, different drugs as well as natural compounds that interfere with proteolysis have been identified and/or developed resulting in homeostatic maintenance and/or the delay of disease progression.
Collapse
Affiliation(s)
- Niki Chondrogianni
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Helenic Research Foundation, 48 Vas. Constantinou Ave., 116 35 Athens, Greece.
| | - Isabelle Petropoulos
- Laboratoire de Biologie Cellulaire du Vieillissement, UR4-UPMC, IFR 83, Université Pierre et Marie Curie-Paris 6, 4 Place Jussieu, 75005 Paris, France
| | - Stefanie Grimm
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich-Schiller University, Dornburger Straße 24, 07743 Jena, Germany
| | - Konstantina Georgila
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Helenic Research Foundation, 48 Vas. Constantinou Ave., 116 35 Athens, Greece
| | - Betul Catalgol
- Department of Biochemistry, Faculty of Medicine, Genetic and Metabolic Diseases Research Center (GEMHAM), Marmara University, Haydarpasa, Istanbul, Turkey
| | - Bertrand Friguet
- Laboratoire de Biologie Cellulaire du Vieillissement, UR4-UPMC, IFR 83, Université Pierre et Marie Curie-Paris 6, 4 Place Jussieu, 75005 Paris, France
| | - Tilman Grune
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich-Schiller University, Dornburger Straße 24, 07743 Jena, Germany
| | - Efstathios S Gonos
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Helenic Research Foundation, 48 Vas. Constantinou Ave., 116 35 Athens, Greece.
| |
Collapse
|
34
|
Watkins GR, Wang N, Mazalouskas MD, Gomez RJ, Guthrie CR, Kraemer BC, Schweiger S, Spiller BW, Wadzinski BE. Monoubiquitination promotes calpain cleavage of the protein phosphatase 2A (PP2A) regulatory subunit α4, altering PP2A stability and microtubule-associated protein phosphorylation. J Biol Chem 2012; 287:24207-15. [PMID: 22613722 DOI: 10.1074/jbc.m112.368613] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Multiple neurodegenerative disorders are linked to aberrant phosphorylation of microtubule-associated proteins (MAPs). Protein phosphatase 2A (PP2A) is the major MAP phosphatase; however, little is known about its regulation at microtubules. α4 binds the PP2A catalytic subunit (PP2Ac) and the microtubule-associated E3 ubiquitin ligase MID1, and through unknown mechanisms can both reduce and enhance PP2Ac stability. We show MID1-dependent monoubiquitination of α4 triggers calpain-mediated cleavage and switches α4's activity from protective to destructive, resulting in increased Tau phosphorylation. This regulatory mechanism appears important in MAP-dependent pathologies as levels of cleaved α4 are decreased in Opitz syndrome and increased in Alzheimer disease, disorders characterized by MAP hypophosphorylation and hyperphosphorylation, respectively. These findings indicate that regulated inter-domain cleavage controls the dual functions of α4, and dysregulation of α4 cleavage may contribute to Opitz syndrome and Alzheimer disease.
Collapse
Affiliation(s)
- Guy R Watkins
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Santa-Maria I, Varghese M, Ksiezak-Reding H, Dzhun A, Wang J, Pasinetti GM. Paired helical filaments from Alzheimer disease brain induce intracellular accumulation of Tau protein in aggresomes. J Biol Chem 2012; 287:20522-33. [PMID: 22496370 DOI: 10.1074/jbc.m111.323279] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Abnormal folding of tau protein leads to the generation of paired helical filaments (PHFs) and neurofibrillary tangles, a key neuropathological feature in Alzheimer disease and tauopathies. A specific anatomical pattern of pathological changes developing in the brain suggests that once tau pathology is initiated it propagates between neighboring neuronal cells, possibly spreading along the axonal network. We studied whether PHFs released from degenerating neurons could be taken up by surrounding cells and promote spreading of tau pathology. Neuronal and non-neuronal cells overexpressing green fluorescent protein-tagged tau (GFP-Tau) were treated with isolated fractions of human Alzheimer disease-derived PHFs for 24 h. We found that cells internalized PHFs through an endocytic mechanism and developed intracellular GFP-Tau aggregates with attributes of aggresomes. This was particularly evident by the perinuclear localization of aggregates and redistribution of the vimentin intermediate filament network and retrograde motor protein dynein. Furthermore, the content of Sarkosyl-insoluble tau, a measure of abnormal tau aggregation, increased 3-fold in PHF-treated cells. An exosome-related mechanism did not appear to be involved in the release of GFP-Tau from untreated cells. The evidence that cells can internalize PHFs, leading to formation of aggresome-like bodies, opens new therapeutic avenues to prevent propagation and spreading of tau pathology.
Collapse
Affiliation(s)
- Ismael Santa-Maria
- Center of Excellence for Novel Approaches to Neurodiagnostics and Neurotherapeutics, Brain Institute, Center of Excellence for Research in Complementary and Alternative Medicine in Alzheimer's Disease, Department of Neurology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | |
Collapse
|
36
|
Aggrephagy: selective disposal of protein aggregates by macroautophagy. Int J Cell Biol 2012; 2012:736905. [PMID: 22518139 PMCID: PMC3320095 DOI: 10.1155/2012/736905] [Citation(s) in RCA: 359] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 01/06/2012] [Indexed: 02/07/2023] Open
Abstract
Protein aggregation is a continuous process in our cells. Some proteins aggregate in a regulated manner required for different vital functional processes in the cells whereas other protein aggregates result from misfolding caused by various stressors. The decision to form an aggregate is largely made by chaperones and chaperone-assisted proteins. Proteins that are damaged beyond repair are degraded either by the proteasome or by the lysosome via autophagy. The aggregates can be degraded by the proteasome and by chaperone-mediated autophagy only after dissolution into soluble single peptide species. Hence, protein aggregates as such are degraded by macroautophagy. The selective degradation of protein aggregates by macroautophagy is called aggrephagy. Here we review the processes of aggregate formation, recognition, transport, and sequestration into autophagosomes by autophagy receptors and the role of aggrephagy in different protein aggregation diseases.
Collapse
|
37
|
Bucci C, Bakke O, Progida C. Charcot-Marie-Tooth disease and intracellular traffic. Prog Neurobiol 2012; 99:191-225. [PMID: 22465036 PMCID: PMC3514635 DOI: 10.1016/j.pneurobio.2012.03.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Revised: 12/23/2011] [Accepted: 03/13/2012] [Indexed: 12/23/2022]
Abstract
Mutations of genes whose primary function is the regulation of membrane traffic are increasingly being identified as the underlying causes of various important human disorders. Intriguingly, mutations in ubiquitously expressed membrane traffic genes often lead to cell type- or organ-specific disorders. This is particularly true for neuronal diseases, identifying the nervous system as the most sensitive tissue to alterations of membrane traffic. Charcot-Marie-Tooth (CMT) disease is one of the most common inherited peripheral neuropathies. It is also known as hereditary motor and sensory neuropathy (HMSN), which comprises a group of disorders specifically affecting peripheral nerves. This peripheral neuropathy, highly heterogeneous both clinically and genetically, is characterized by a slowly progressive degeneration of the muscle of the foot, lower leg, hand and forearm, accompanied by sensory loss in the toes, fingers and limbs. More than 30 genes have been identified as targets of mutations that cause CMT neuropathy. A number of these genes encode proteins directly or indirectly involved in the regulation of intracellular traffic. Indeed, the list of genes linked to CMT disease includes genes important for vesicle formation, phosphoinositide metabolism, lysosomal degradation, mitochondrial fission and fusion, and also genes encoding endosomal and cytoskeletal proteins. This review focuses on the link between intracellular transport and CMT disease, highlighting the molecular mechanisms that underlie the different forms of this peripheral neuropathy and discussing the pathophysiological impact of membrane transport genetic defects as well as possible future ways to counteract these defects.
Collapse
Affiliation(s)
- Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni, 73100 Lecce, Italy.
| | | | | |
Collapse
|
38
|
D'Eletto M, Farrace MG, Rossin F, Strappazzon F, Giacomo GD, Cecconi F, Melino G, Sepe S, Moreno S, Fimia GM, Falasca L, Nardacci R, Piacentini M. Type 2 transglutaminase is involved in the autophagy-dependent clearance of ubiquitinated proteins. Cell Death Differ 2012; 19:1228-38. [PMID: 22322858 DOI: 10.1038/cdd.2012.2] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Eukaryotic cells are equipped with an efficient quality control system to selectively eliminate misfolded and damaged proteins, and organelles. Abnormal polypeptides that escape from proteasome-dependent degradation and aggregate in the cytosol can be transported via microtubules to inclusion bodies called 'aggresomes', where misfolded proteins are confined and degraded by autophagy. Here, we show that Type 2 transglutaminase (TG2) knockout mice display impaired autophagy and accumulate ubiquitinated protein aggregates upon starvation. Furthermore, p62-dependent peroxisome degradation is also impaired in the absence of TG2. We also demonstrate that, under cellular stressful conditions, TG2 physically interacts with p62 and they are localized in cytosolic protein aggregates, which are then recruited into autophagosomes, where TG2 is degraded. Interestingly, the enzyme's crosslinking activity is activated during autophagy and its inhibition leads to the accumulation of ubiquitinated proteins. Taken together, these data indicate that the TG2 transamidating activity has an important role in the assembly of protein aggregates, as well as in the clearance of damaged organelles by macroautophagy.
Collapse
Affiliation(s)
- M D'Eletto
- Department of Biology, University of Rome 'Tor Vergata', Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Trüe O, Matthias P. Interplay between histone deacetylases and autophagy--from cancer therapy to neurodegeneration. Immunol Cell Biol 2011; 90:78-84. [PMID: 22124372 DOI: 10.1038/icb.2011.103] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Histone deacetylases (HDACs) are chromatin modifiers that alter gene expression but also exert a broad range of functions outside the nucleus by deacetylating non-histone target proteins. They gained growing attention for their implications in disease treatment, mainly through research using HDAC-inhibiting compounds. Understanding the effects of HDAC function and deregulation has therefore become an important focus for both basic and applied research. One of the described effects of HDAC inhibition is induction of autophagy. Autophagy is a ubiquitous process of recycling cellular components in response to starvation or stress and therefore crucial for cell homeostasis. Because of its role in managing anomalous protein overloads, autophagy is of great interest for neurodegenerative disease research. However, autophagy can also promote cell death, which puts it in the focus of cancer research. This review provides an overview of what we know of the impact of HDACs on the autophagy pathway and describes the fields where promising progress has been achieved, although one has to state that the work to illuminate the connections has just begun. Therefore, one focus is the effect of HDAC inhibition on autophagy in several types and models of cancer, which aims to find combinations of treatments that circumvent the ability of cancer cells to escape from cell death. Another recently emerged aspect is the direct involvement of the cytosolic deacetylase HDAC6 in autophagy progression, which is of great potential for revealing disease mechanisms in neurodegeneration.
Collapse
Affiliation(s)
- Oliver Trüe
- Friedrich Miescher Institute For Biomedical Research, Basel, Switzerland
| | | |
Collapse
|
40
|
Guthrie CR, Greenup L, Leverenz JB, Kraemer BC. MSUT2 is a determinant of susceptibility to tau neurotoxicity. Hum Mol Genet 2011; 20:1989-99. [PMID: 21355046 DOI: 10.1093/hmg/ddr079] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lesions containing abnormal aggregated tau protein are one of the diagnostic hallmarks of Alzheimer's disease (AD) and related tauopathy disorders. How aggregated tau leads to dementia remains enigmatic, although neuronal dysfunction and loss clearly contribute. We previously identified sut-2 as a gene required for tau neurotoxicity in a transgenic Caenorhabditis elegans model of tauopathy. Here, we further explore the role of sut-2 and show that overexpression of SUT-2 protein enhances tau-induced neuronal dysfunction, neurotoxicity and accumulation of insoluble tau. We also explore the relationship between sut-2 and its human homolog, mammalian SUT-2 (MSUT2) and find both proteins to be predominantly nuclear and localized to SC35-positive nuclear speckles. Using a cell culture model for the accumulation of pathological tau, we find that high tau levels lead to increased expression of MSUT2 protein. We analyzed MSUT2 protein in age-matched post-mortem brain samples from AD patients and observe a marked decrease in overall MSUT2 levels in the temporal lobe of AD patients. Analysis of post-mortem tissue from AD cases shows a clear reduction in neuronal MSUT2 levels in brain regions affected by tau pathology, but little change in regions lacking tau pathology. RNAi knockdown of MSUT2 in cultured human cells overexpressing tau causes a marked decrease in tau aggregation. Both cell culture and post-mortem tissue studies suggest that MSUT2 levels may influence neuronal vulnerability to tau toxicity and aggregation. Thus, neuroprotective strategies targeting MSUT2 may be of therapeutic interest for tauopathy disorders.
Collapse
Affiliation(s)
- Chris R Guthrie
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle Division, Seattle, WA 98108, USA
| | | | | | | |
Collapse
|