1
|
Wang X, Li C, Chen L, He B, Li Y. Therapeutic potential of dual HDAC6/SIRT2 inhibition in Alzheimer's disease. Eur J Med Chem 2025; 294:117733. [PMID: 40381221 DOI: 10.1016/j.ejmech.2025.117733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/25/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by hallmark pathological changes such as amyloid β (Aβ) plaques, neurofibrillary tangles (NFTs) due to tau hyperphosphorylation, and neuroinflammation. Current therapeutic approaches focusing on single-target strategies exhibit limited efficacy, necessitating the exploration of novel multi-target approaches. Histone deacetylase 6 (HDAC6) and SIRT2, as two types of cytosolic histone deacetylases, have emerged as promising targets for AD treatment. HDAC6 plays a role in tau protein phosphorylation, while SIRT2 is involved in Aβ production. Both enzymes regulate microtubule proteins, impacting the formation of NFTs and Aβ plaques. Inhibition of HDAC6 reduces tau hyperphosphorylation, improves microtubule stability, and mitigates neuroinflammation, whereas SIRT2 inhibition attenuates Aβ accumulation and neuroinflammation. Recent studies indicate that dual-targeted inhibition of HDAC6 and SIRT2 may exhibit synergistic effects, suggesting it as a promising strategy for AD treatment. This review summarizes the biological roles of HDAC6 and SIRT2 in AD pathology and examines the development of dual-target inhibitors. It also discusses the challenges, including selectivity and toxicity, emphasizing that the development of combined HDAC6 and SIRT2 inhibitors represents a new direction for future AD treatment.
Collapse
Affiliation(s)
- Xingyu Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China
| | - Cunjiang Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China
| | - Lei Chen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China
| | - Bin He
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China.
| | - Yan Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China; School of Basic Medical Science, Guizhou Medical University, Guian New Area, 561113, China.
| |
Collapse
|
2
|
Ma Y, Wang W, Liu S, Qiao X, Xing Y, Zhou Q, Zhang Z. Epigenetic Regulation of Neuroinflammation in Alzheimer's Disease. Cells 2023; 13:79. [PMID: 38201283 PMCID: PMC10778497 DOI: 10.3390/cells13010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Alzheimer's disease (AD) is a chronic and progressive neurodegenerative disease and clinically manifests with cognitive decline and behavioral disabilities. Over the past years, mounting studies have demonstrated that the inflammatory response plays a key role in the onset and development of AD, and neuroinflammation has been proposed as the third major pathological driving factor of AD, ranking after the two well-known core pathologies, amyloid β (Aβ) deposits and neurofibrillary tangles (NFTs). Epigenetic mechanisms, referring to heritable changes in gene expression independent of DNA sequence alterations, are crucial regulators of neuroinflammation which have emerged as potential therapeutic targets for AD. Upon regulation of transcriptional repression or activation, epigenetic modification profiles are closely involved in inflammatory gene expression and signaling pathways of neuronal differentiation and cognitive function in central nervous system disorders. In this review, we summarize the current knowledge about epigenetic control mechanisms with a focus on DNA and histone modifications involved in the regulation of inflammatory genes and signaling pathways in AD, and the inhibitors under clinical assessment are also discussed.
Collapse
Affiliation(s)
- Yajing Ma
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China;
| | - Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.W.); (Y.X.)
| | - Sufang Liu
- Department of Biomedical Sciences, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA;
| | - Xiaomeng Qiao
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China;
| | - Ying Xing
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.W.); (Y.X.)
| | - Qingfeng Zhou
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China;
| | - Zhijian Zhang
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China;
| |
Collapse
|
3
|
Silvestro S, Raffaele I, Mazzon E. Modulating Stress Proteins in Response to Therapeutic Interventions for Parkinson's Disease. Int J Mol Sci 2023; 24:16233. [PMID: 38003423 PMCID: PMC10671288 DOI: 10.3390/ijms242216233] [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: 10/03/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative illness characterized by the degeneration of dopaminergic neurons in the substantia nigra, resulting in motor symptoms and without debilitating motors. A hallmark of this condition is the accumulation of misfolded proteins, a phenomenon that drives disease progression. In this regard, heat shock proteins (HSPs) play a central role in the cellular response to stress, shielding cells from damage induced by protein aggregates and oxidative stress. As a result, researchers have become increasingly interested in modulating these proteins through pharmacological and non-pharmacological therapeutic interventions. This review aims to provide an overview of the preclinical experiments performed over the last decade in this research field. Specifically, it focuses on preclinical studies that center on the modulation of stress proteins for the treatment potential of PD. The findings display promise in targeting HSPs to ameliorate PD outcomes. Despite the complexity of HSPs and their co-chaperones, proteins such as HSP70, HSP27, HSP90, and glucose-regulated protein-78 (GRP78) may be efficacious in slowing or preventing disease progression. Nevertheless, clinical validation is essential to confirm the safety and effectiveness of these preclinical approaches.
Collapse
Affiliation(s)
| | | | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (I.R.)
| |
Collapse
|
4
|
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
|
5
|
Çetin Ö, Sari S, Erdem-Yurter H, Bora G. Rutin increases alpha-tubulin acetylation via histone deacetylase 6 inhibition. Drug Dev Res 2022; 83:993-1002. [PMID: 35266183 DOI: 10.1002/ddr.21927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/04/2022] [Accepted: 02/10/2022] [Indexed: 11/11/2022]
Abstract
Microtubules are dynamic cytoskeletal filaments composed of alpha- (α) and beta (β)-tubulin proteins. α-tubulin proteins are posttranslationally acetylated, and loss of acetylation is associated with axonal transport defects, a common alteration contributing to the pathomechanisms of several neurodegenerative diseases. Restoring α-tubulin acetylation by pharmacological inhibition of HDAC6, a primary α-tubulin deacetylase, can rescue impaired transport. Therefore, HDAC6 is considered a promising therapeutic target for neurodegenerative diseases, but currently, there is no clinically approved inhibitor for this purpose. In this study, using drug repurposing strategy, we aimed to identify compounds possessing HDAC6 inhibition activity and inducing α-tubulin acetylation. We systematically analyzed the FDA-approved library by utilizing virtual screening and consensus scoring approaches. Inhibition activities of promising compounds were tested using in vitro assays. Motor neuron-like NSC34 cells were treated with the candidate compounds, and α-tubulin acetylation levels were determined by Western blot. Our results demonstrated that rutin, a natural flavonoid, inhibits cellular HDAC6 activity without inducing any toxicity, and it significantly increases α-tubulin acetylation level in motor neuron-like cells.
Collapse
Affiliation(s)
- Özge Çetin
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.,The Faculty of Health and Medical Sciences, School of Medicine, Keele University, Staffordshire, UK
| | - Suat Sari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Hayat Erdem-Yurter
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Gamze Bora
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
6
|
Blaudin de Thé FX, Lassus B, Schaler AW, Fowler SL, Goulbourne CN, Jeggo R, Mannoury la Cour C, Millan MJ, Duff KE. P62 accumulates through neuroanatomical circuits in response to tauopathy propagation. Acta Neuropathol Commun 2021; 9:177. [PMID: 34727983 PMCID: PMC8561893 DOI: 10.1186/s40478-021-01280-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/17/2021] [Indexed: 12/26/2022] Open
Abstract
In Alzheimer's disease and related tauopathies, trans-synaptic transfer and accumulation of pathological tau from donor to recipient neurons is thought to contribute to disease progression, but the underlying mechanisms are poorly understood. Using complementary in vivo and in vitro models, we examined the relationship between these two processes and neuronal clearance. Accumulation of p62 (a marker of defective protein clearance) correlated with pathological tau accumulation in two mouse models of tauopathy spread; Entorhinal Cortex-tau (EC-Tau) mice where tau pathology progresses in time from EC to other brain regions, and PS19 mice injected with tau seeds. In both models and in several brain regions, p62 colocalized with human tau in a pathological conformation (MC1 antibody). In EC-Tau mice, p62 accumulated before overt tau pathology had developed and was associated with the presence of aggregation-competent tau seeds identified using a FRET-based assay. Furthermore, p62 accumulated in the cytoplasm of neurons in the dentate gyrus of EC-Tau mice prior to the appearance of MC1 positive tauopathy. However, MC1 positive tau was shown to be present at the synapse and to colocalize with p62 as shown by immuno electron microscopy. In vitro, p62 colocalized with tau inclusions in two primary cortical neuron models of tau pathology. In a three-chamber microfluidic device containing neurons overexpressing fluorescent tau, seeding of tau in the donor chamber led to tau pathology spread and p62 accumulation in both the donor and the recipient chamber. Overall, these data are in accordance with the hypothesis that the accumulation and trans-synaptic spread of pathological tau disrupts clearance mechanisms, preceding the appearance of obvious tau aggregation. A vicious cycle of tau accumulation and clearance deficit would be expected to feed-forward and exacerbate disease progression across neuronal circuits in human tauopathies.
Collapse
Affiliation(s)
- François-Xavier Blaudin de Thé
- Taub Institute for Research On Alzheimer’s Disease and the Aging Brain, 630 W 168th St, NY 10032 New York, USA
- Neuroscience and Immunoinflammation Therapeutic Area, Institut de Recherche Servier, 125 Chemin de Ronde, 78290 Croissy-sur-Seine, France
| | - Benjamin Lassus
- Taub Institute for Research On Alzheimer’s Disease and the Aging Brain, 630 W 168th St, NY 10032 New York, USA
- HiFiBiO Therapeutics Pépinière Paris Santé Cochin, 29 Rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Ari W. Schaler
- Taub Institute for Research On Alzheimer’s Disease and the Aging Brain, 630 W 168th St, NY 10032 New York, USA
- UCLA – Physiology Department, 10833 Le Conte Ave, CHS76200, CA 90095 Los Angeles, USA
| | - Stephanie L. Fowler
- Taub Institute for Research On Alzheimer’s Disease and the Aging Brain, 630 W 168th St, NY 10032 New York, USA
- UK Dementia Research Institute at UCL, University College London, 90 Gower St, W1T 7NF London, UK
| | - Chris N. Goulbourne
- Nathan Kline Institute: Center for Dementia Research, 140 Old Orangeburg Road, NY 10962 Orangeburg, USA
| | - Ross Jeggo
- Neuroscience and Immunoinflammation Therapeutic Area, Institut de Recherche Servier, 125 Chemin de Ronde, 78290 Croissy-sur-Seine, France
| | - Clotilde Mannoury la Cour
- Neuroscience and Immunoinflammation Therapeutic Area, Institut de Recherche Servier, 125 Chemin de Ronde, 78290 Croissy-sur-Seine, France
| | - Mark J. Millan
- Neuroscience and Immunoinflammation Therapeutic Area, Institut de Recherche Servier, 125 Chemin de Ronde, 78290 Croissy-sur-Seine, France
- Present Address: Institute of Neuroscience and Psychology, College of Medicine, Vet and Life Science, Glasgow University, Glasgow, G12 8QQ United Kingdom
| | - Karen E. Duff
- Taub Institute for Research On Alzheimer’s Disease and the Aging Brain, 630 W 168th St, NY 10032 New York, USA
- UK Dementia Research Institute at UCL, University College London, 90 Gower St, W1T 7NF London, UK
| |
Collapse
|
7
|
Singh A, Yadav A, Phogat J, Dabur R. Dynamics of autophagy and ubiquitin proteasome system coordination and interplay in skeletal muscle atrophy. Curr Mol Pharmacol 2021; 15:475-486. [PMID: 34365963 DOI: 10.2174/1874467214666210806163851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/26/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
Skeletal muscles are considered the largest reservoirs of the protein pool in the body and are critical for the maintenances of body homeostasis. Skeletal muscle atrophy is supported by various physiopathological conditions that lead to loss of muscle mass and contractile capacity of the skeletal muscle. Lysosomal mediated autophagy and ubiquitin-proteasomal system (UPS) concede the major intracellular systems of muscle protein degradation that result in the loss of mass and strength. Both systems recognize ubiquitination as a signal of degradation through different mechanisms, a sign of dynamic interplay between systems. Hence, growing shreds of evidence suggest the interdependency of autophagy and UPS in the progression of skeletal muscle atrophy under various pathological conditions. Therefore, understanding the molecular dynamics as well associated factors responsible for their interdependency is a necessity for the new therapeutic insights to counteract the muscle loss. Based on current literature, the present review summarizes the factors interplay in between the autophagy and UPS in favor of enhanced proteolysis of skeletal muscle and how they affect the anabolic signaling pathways under various conditions of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Ajay Singh
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Aarti Yadav
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Jatin Phogat
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Rajesh Dabur
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| |
Collapse
|
8
|
Cenini G, Hebisch M, Iefremova V, Flitsch LJ, Breitkreuz Y, Tanzi RE, Kim DY, Peitz M, Brüstle O. Dissecting Alzheimer's disease pathogenesis in human 2D and 3D models. Mol Cell Neurosci 2021; 110:103568. [DOI: 10.1016/j.mcn.2020.103568] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
|
9
|
LoPresti P. HDAC6 in Diseases of Cognition and of Neurons. Cells 2020; 10:E12. [PMID: 33374719 PMCID: PMC7822434 DOI: 10.3390/cells10010012] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) neurodegenerative diseases are characterized by faulty intracellular transport, cognition, and aggregate regulation. Traditionally, neuroprotection exerted by histone deacetylase (HDAC) inhibitors (HDACi) has been attributed to the ability of this drug class to promote histone acetylation. However, HDAC6 in the healthy CNS functions via distinct mechanisms, due largely to its cytoplasmic localization. Indeed, in healthy neurons, cytoplasmic HDAC6 regulates the acetylation of a variety of non-histone proteins that are linked to separate functions, i.e., intracellular transport, neurotransmitter release, and aggregate formation. These three HDAC6 activities could work independently or in synergy. Of particular interest, HDAC6 targets the synaptic protein Bruchpilot and neurotransmitter release. In pathological conditions, HDAC6 becomes abundant in the nucleus, with deleterious consequences for transcription regulation and synapses. Thus, HDAC6 plays a leading role in neuronal health or dysfunction. Here, we review recent findings and novel conclusions on the role of HDAC6 in neurodegeneration. Selective studies with pan-HDACi are also included. We propose that an early alteration of HDAC6 undermines synaptic transmission, while altering transport and aggregation, eventually leading to neurodegeneration.
Collapse
Affiliation(s)
- Patrizia LoPresti
- Department of Psychology, University of Illinois at Chicago, 1007 West Harrison Street, Chicago, IL 60607, USA
| |
Collapse
|
10
|
Trzeciakiewicz H, Ajit D, Tseng JH, Chen Y, Ajit A, Tabassum Z, Lobrovich R, Peterson C, Riddick NV, Itano MS, Tripathy A, Moy SS, Lee VMY, Trojanowski JQ, Irwin DJ, Cohen TJ. An HDAC6-dependent surveillance mechanism suppresses tau-mediated neurodegeneration and cognitive decline. Nat Commun 2020; 11:5522. [PMID: 33139698 PMCID: PMC7606452 DOI: 10.1038/s41467-020-19317-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/08/2020] [Indexed: 01/15/2023] Open
Abstract
Tauopathies including Alzheimer's disease (AD) are marked by the accumulation of aberrantly modified tau proteins. Acetylated tau, in particular, has recently been implicated in neurodegeneration and cognitive decline. HDAC6 reversibly regulates tau acetylation, but its role in tauopathy progression remains unclear. Here, we identified an HDAC6-chaperone complex that targets aberrantly modified tau. HDAC6 not only deacetylates tau but also suppresses tau hyperphosphorylation within the microtubule-binding region. In neurons and human AD brain, HDAC6 becomes co-aggregated within focal tau swellings and human AD neuritic plaques. Using mass spectrometry, we identify a novel HDAC6-regulated tau acetylation site as a disease specific marker for 3R/4R and 3R tauopathies, supporting uniquely modified tau species in different neurodegenerative disorders. Tau transgenic mice lacking HDAC6 show reduced survival characterized by accelerated tau pathology and cognitive decline. We propose that a HDAC6-dependent surveillance mechanism suppresses toxic tau accumulation, which may protect against the progression of AD and related tauopathies.
Collapse
Affiliation(s)
- Hanna Trzeciakiewicz
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599-7260, USA
| | - Deepa Ajit
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599-7250, USA
| | - Jui-Heng Tseng
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599-7250, USA
| | - Youjun Chen
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599-7250, USA
| | - Aditi Ajit
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599-7250, USA
| | - Zarin Tabassum
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599-7250, USA
| | - Rebecca Lobrovich
- Penn Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-4283, USA
| | - Claire Peterson
- Penn Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-4283, USA
| | - Natallia V Riddick
- Division of Comparative Medicine, University of North Carolina, Chapel Hill, NC, 27599-7146, USA
| | - Michelle S Itano
- Department of Cell Biology and Physiology, Carolina Institute for Developmental Disabilities, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599-7250, USA
| | - Ashutosh Tripathy
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599-7260, USA
| | - Sheryl S Moy
- Department of Psychiatry, Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, 27599-7146, USA
| | - Virginia M Y Lee
- Department of Pathology & Laboratory Medicine, Center for Neurodegenerative Disease Research (CNDR), Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-4283, USA
| | - John Q Trojanowski
- Department of Pathology & Laboratory Medicine, Center for Neurodegenerative Disease Research (CNDR), Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-4283, USA
| | - David J Irwin
- Penn Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-4283, USA
| | - Todd J Cohen
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599-7260, USA.
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599-7250, USA.
| |
Collapse
|
11
|
Kulka LAM, Fangmann PV, Panfilova D, Olzscha H. Impact of HDAC Inhibitors on Protein Quality Control Systems: Consequences for Precision Medicine in Malignant Disease. Front Cell Dev Biol 2020; 8:425. [PMID: 32582706 PMCID: PMC7291789 DOI: 10.3389/fcell.2020.00425] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/07/2020] [Indexed: 12/21/2022] Open
Abstract
Lysine acetylation is one of the major posttranslational modifications (PTM) in human cells and thus needs to be tightly regulated by the writers of this process, the histone acetyl transferases (HAT), and the erasers, the histone deacetylases (HDAC). Acetylation plays a crucial role in cell signaling, cell cycle control and in epigenetic regulation of gene expression. Bromodomain (BRD)-containing proteins are readers of the acetylation mark, enabling them to transduce the modification signal. HDAC inhibitors (HDACi) have been proven to be efficient in hematologic malignancies with four of them being approved by the FDA. However, the mechanisms by which HDACi exert their cytotoxicity are only partly resolved. It is likely that HDACi alter the acetylation pattern of cytoplasmic proteins, contributing to their anti-cancer potential. Recently, it has been demonstrated that various protein quality control (PQC) systems are involved in recognizing the altered acetylation pattern upon HDACi treatment. In particular, molecular chaperones, the ubiquitin proteasome system (UPS) and autophagy are able to sense the structurally changed proteins, providing additional targets. Recent clinical studies of novel HDACi have proven that proteins of the UPS may serve as biomarkers for stratifying patient groups under HDACi regimes. In addition, members of the PQC systems have been shown to modify the epigenetic readout of HDACi treated cells and alter proteostasis in the nucleus, thus contributing to changing gene expression profiles. Bromodomain (BRD)-containing proteins seem to play a potent role in transducing the signaling process initiating apoptosis, and many clinical trials are under way to test BRD inhibitors. Finally, it has been demonstrated that HDACi treatment leads to protein misfolding and aggregation, which may explain the effect of panobinostat, the latest FDA approved HDACi, in combination with the proteasome inhibitor bortezomib in multiple myeloma. Therefore, proteins of these PQC systems provide valuable targets for precision medicine in cancer. In this review, we give an overview of the impact of HDACi treatment on PQC systems and their implications for malignant disease. We exemplify the development of novel HDACi and how affected proteins belonging to PQC can be used to determine molecular signatures and utilized in precision medicine.
Collapse
Affiliation(s)
- Linda Anna Michelle Kulka
- Medical Faculty, Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Pia-Victoria Fangmann
- Medical Faculty, Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Diana Panfilova
- Medical Faculty, Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Heidi Olzscha
- Medical Faculty, Institute of Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
12
|
Cao XY, Liu J, Zhang YJ, Wang Y, Xiong JW, Wu J, Chen L. Exposure of adult mice to perfluorobutanesulfonate impacts ovarian functions through hypothyroxinemia leading to down-regulation of Akt-mTOR signaling. CHEMOSPHERE 2020; 244:125497. [PMID: 31809938 DOI: 10.1016/j.chemosphere.2019.125497] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 06/10/2023]
Abstract
Perfluorobutanesulfonate (PFBS), a short-chain perfluoroalkyl substance, is used in many industrial products. Preliminary evidence suggests that exposure to PFBS may increase the risk of infertility. The aim of this study was to investigate the influence of PFBS on ovarian function. Herein, we show that exposure of adult female mice to PFBS (200 mg/kg/day) (PFBS-mice) caused a decrease in the levels of serum total triiodothyronine and thyroxine, which depended on the activation of peroxisome proliferator-activated receptor α (PPARα). The numbers of secondary, early antral and antral follicles were reduced in PFBS-mice with an increase in the atretic follicles, and these changes were recovered by the replacement of L-thyroxinein or the treatment with PPARα antagonist GW6471. PFBS-induced hypothyroxinemia led to a decrease in the levels of Akt, mTOR and p70S6K phosphorylation in ovarian granular cells and cumulus cells, which suppressed the proliferation of these cells and enhanced autophagic death of granular cells and cumulus cells. The levels of serum estradiol and progesterone were reduced in PFBS-mice with a low expression of the steroidogenic genes Star and P450scc in ovarian tissues, which were sensitive to the replacement of L-thyroxinein or the blockade of PPARα. The results indicate that exposure to PFBS (≥200 mg/kg/day) through reducing thyroid hormones causes down-regulation of Akt-mTOR signaling in granular cells and cumulus cells, leading to the deficits in the development of follicles and the biosynthesis of ovarian hormones.
Collapse
Affiliation(s)
- Xin-Yuan Cao
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China; Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Juan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China; Yangzhou Municipal Maternal and Child Health Hospital, Yangzhou, Jiangsu, 225001, China
| | - Ya-Jie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Jian-Wei Xiong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jie Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Ling Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China; Department of Physiology, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
13
|
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
|
14
|
Kareem O, Bader GN, Pottoo FH, Amir M, Barkat MA, Pandey M. Beclin 1 Complex and Neurodegenerative Disorders. QUALITY CONTROL OF CELLULAR PROTEIN IN NEURODEGENERATIVE DISORDERS 2020. [DOI: 10.4018/978-1-7998-1317-0.ch009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Beclin1 is the mammalian orthologue of yeast Atg6/vacuolar protein sorting-30 (VPS30). Beclin1 interacts with various biological macromolecules like ATG14, BIF-1, NRBF2, RUBICON, UVRAG, AMBRA1, HMGB1, PINK1, and PARKIN. Such interactions promote Beclin1-PI3KC3 complex formation. Autophagy is blocked in apoptosis owing to the breakdown of Beclin1 by caspase whereas autophagy induction inhibits effector caspase degradation, therefore, blocks apoptosis. Thus, the Beclin1 is an essential biomolecular species for cross-regulation between autophagy and apoptosis. Various studies carried out in neurodegenerative animal models associated with aggregated proteins have confirmed that multifunctional Beclin1 protein is necessary for neuronal integrity. The role of Beclin1 protein has been investigated and was reported in various human neurodegeneration disorders. This chapter aims to provide an insight into the role of Beclin1 in the development of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ozaifa Kareem
- Department of Pharmaceutical Sciences (Pharmacology Division), Faculty of Applied Sciences and Technology, University of Kashmir, Srinagar, India
| | - Ghulam Nabi Bader
- Department of Pharmaceutical Sciences (Pharmacology Division), Faculty of Applied Sciences and Technology, University of Kashmir, Srinagar, India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Saudi Arabia
| | - Mohd. Amir
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Saudi Arabia
| | - Md. Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al-Batin, Saudi Arabia
| | - Mukesh Pandey
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, India
| |
Collapse
|
15
|
Kuta R, Larochelle N, Fernandez M, Pal A, Minotti S, Tibshirani M, St Louis K, Gentil BJ, Nalbantoglu JN, Hermann A, Durham HD. Depending on the stress, histone deacetylase inhibitors act as heat shock protein co-inducers in motor neurons and potentiate arimoclomol, exerting neuroprotection through multiple mechanisms in ALS models. Cell Stress Chaperones 2020; 25:173-191. [PMID: 31900865 PMCID: PMC6985055 DOI: 10.1007/s12192-019-01064-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/12/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Upregulation of heat shock proteins (HSPs) is an approach to treatment of neurodegenerative disorders with impaired proteostasis. Many neurons, including motor neurons affected in amyotrophic lateral sclerosis (ALS), are relatively resistant to stress-induced upregulation of HSPs. This study demonstrated that histone deacetylase (HDAC) inhibitors enable the heat shock response in cultured spinal motor neurons, in a stress-dependent manner, and can improve the efficacy of HSP-inducing drugs in murine spinal cord cultures subjected to thermal or proteotoxic stress. The effect of particular HDAC inhibitors differed with the stress paradigm. The HDAC6 (class IIb) inhibitor, tubastatin A, acted as a co-inducer of Hsp70 (HSPA1A) expression with heat shock, but not with proteotoxic stress induced by expression of mutant SOD1 linked to familial ALS. Certain HDAC class I inhibitors (the pan inhibitor, SAHA, or the HDAC1/3 inhibitor, RGFP109) were HSP co-inducers comparable to the hydroxyamine arimoclomol in response to proteotoxic stress, but not thermal stress. Regardless, stress-induced Hsp70 expression could be enhanced by combining an HDAC inhibitor with either arimoclomol or with an HSP90 inhibitor that constitutively induced HSPs. HDAC inhibition failed to induce Hsp70 in motor neurons expressing ALS-linked mutant FUS, in which the heat shock response was suppressed; yet SAHA, RGFP109, and arimoclomol did reduce loss of nuclear FUS, a disease hallmark, and HDAC inhibition rescued the DNA repair response in iPSC-derived motor neurons carrying the FUSP525Lmutation, pointing to multiple mechanisms of neuroprotection by both HDAC inhibiting drugs and arimoclomol.
Collapse
Affiliation(s)
- Rachel Kuta
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Nancy Larochelle
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Mario Fernandez
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Arun Pal
- Department Neurology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Sandra Minotti
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Michael Tibshirani
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Kyle St Louis
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Benoit J Gentil
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Josephine N Nalbantoglu
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht-Kossel", Department of Neurology and Center for Transdisciplinary Neuroscience (CTNR), University Medical Center Rostock, University of Rostock, Rostock, Germany and German Center for Neurodegenerative Diseases (DZNE) Rostock, Rostock, Germany
| | - Heather D Durham
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada.
| |
Collapse
|
16
|
Screening of a neuronal cell model of tau pathology for therapeutic compounds. Neurobiol Aging 2019; 76:24-34. [DOI: 10.1016/j.neurobiolaging.2018.11.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/23/2018] [Accepted: 11/30/2018] [Indexed: 11/20/2022]
|
17
|
Iaconelli J, Xuan L, Karmacharya R. HDAC6 Modulates Signaling Pathways Relevant to Synaptic Biology and Neuronal Differentiation in Human Stem-Cell-Derived Neurons. Int J Mol Sci 2019; 20:ijms20071605. [PMID: 30935091 PMCID: PMC6480207 DOI: 10.3390/ijms20071605] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 12/18/2022] Open
Abstract
Recent studies show that histone deacetylase 6 (HDAC6) has important roles in the human brain, especially in the context of a number of nervous system disorders. Animal models of neurodevelopmental, neurodegenerative, and neuropsychiatric disorders show that HDAC6 modulates important biological processes relevant to disease biology. Pan-selective histone deacetylase (HDAC) inhibitors had been studied in animal behavioral assays and shown to induce synaptogenesis in rodent neuronal cultures. While most studies of HDACs in the nervous system have focused on class I HDACs located in the nucleus (e.g., HDACs 1,2,3), recent findings in rodent models suggest that the cytoplasmic class IIb HDAC, HDAC6, plays an important role in regulating mood-related behaviors. Human studies suggest a significant role for synaptic dysfunction in the prefrontal cortex (PFC) and hippocampus in depression. Studies of HDAC inhibitors (HDACi) in human neuronal cells show that HDAC6 inhibitors (HDAC6i) increase the acetylation of specific lysine residues in proteins involved in synaptogenesis. This has led to the hypothesis that HDAC6i may modulate synaptic biology not through effects on the acetylation of histones, but by regulating acetylation of non-histone proteins.
Collapse
Affiliation(s)
- Jonathan Iaconelli
- Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA.
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Lucius Xuan
- Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA.
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Harvard Medical School and Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA.
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
- Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA 02478, USA.
- Program in Neuroscience, Harvard University, Cambridge, MA 02138, USA.
- Chemical Biology PhD Program, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
18
|
Chang H, Di T, Wang Y, Zeng X, Li G, Wan Q, Yu W, Chen L. Seipin deletion in mice enhances phosphorylation and aggregation of tau protein through reduced neuronal PPARγ and insulin resistance. Neurobiol Dis 2019; 127:350-361. [PMID: 30910747 DOI: 10.1016/j.nbd.2019.03.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/23/2019] [Accepted: 03/21/2019] [Indexed: 12/31/2022] Open
Abstract
Congenital generalized lipodystrophy 2 (CGL2) is characterized by loss of adipose tissue, insulin resistance and cognitive deficits and caused by mutation of BSCL2/seipin gene. Seipin deletion in mice and rats causes severe lipodystrophy, insulin resistance, and cognitive impairment. Hippocampal neurons express seipin protein. This study aimed to investigate the influence of systemic seipin knockout (seipin-sKO), neuronal seipin knockout (seipin-nKO) or adipose seipin knockout (seipin-aKO) in hippocampal tau phosphorylation and aggregation. Levels of tau phosphorylation at Thr212/Ser214 and Ser202/Thr205 and oligomer tau protein were increased in seipin-sKO mice and seipin-nKO mice with a decrease in axonal density and expression of PPARγ. Neuronal seipin deletion increased activities of GSK3β and Akt/mTOR signaling, which were corrected by the administration of PPARγ agonist rosiglitazone for 7 days. The autophagosome formation was reduced in seipin-sKO mice and seipin-nKO mice, which was rescued by the Akt and mTOR inhibitors. The administration of rosiglitazone or Akt, mTOR and GSK3β inhibitors for 7 days could correct the hyperphosphorylation and aggregation of tau. On the other hand, seipin-sKO mice appeared insulin resistance and an increase in phosphorylation of tau at Ser396 and JNK, which were corrected by treatment with rosiglitazone for 30 days rather than 7 days. Inhibition of JNK in seipin-sKO mice corrected the hyperphosphorylated tau at Ser396. The results indicate that neuronal seipin deletion causes hyperphosphorylation and aggregation of tau protein leading to axonal atrophy through reduced PPARγ to enhance GSK3β and Akt/mTOR signaling; systemic seipin deletion-induced insulin resistance causes tau hyperphosphorylation via cascading JNK pathway.
Collapse
Affiliation(s)
- Huanxian Chang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China; Department of Neurology, First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, China
| | - Tingting Di
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China; Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Xianying Zeng
- Department of Geratology, First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, China
| | - Guoxi Li
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Qi Wan
- Department of Neurology, First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, China
| | - Wenfeng Yu
- Key Laboratory of Endemic and Ethnic Diseases of Education Ministry, Guizhou Medical University, Guian New District, Guizhou 550025, China.
| | - Ling Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China; Department of Physiology, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
19
|
Autophagy and Ubiquitin-Proteasome System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1206:527-550. [DOI: 10.1007/978-981-15-0602-4_25] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
20
|
Casares-Crespo L, Calatayud-Baselga I, García-Corzo L, Mira H. On the Role of Basal Autophagy in Adult Neural Stem Cells and Neurogenesis. Front Cell Neurosci 2018; 12:339. [PMID: 30349462 PMCID: PMC6187079 DOI: 10.3389/fncel.2018.00339] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/13/2018] [Indexed: 12/31/2022] Open
Abstract
Adult neurogenesis persists in the adult mammalian brain due to the existence of neural stem cell (NSC) reservoirs in defined niches, where they give rise to new neurons throughout life. Recent research has begun to address the implication of constitutive (basal) autophagy in the regulation of neurogenesis in the mature brain. This review summarizes the current knowledge on the role of autophagy-related genes in modulating adult NSCs, progenitor cells and their differentiation into neurons. The general function of autophagy in neurogenesis in several areas of the embryonic forebrain is also revisited. During development, basal autophagy regulates Wnt and Notch signaling and is mainly required for adequate neuronal differentiation. The available data in the adult indicate that the autophagy-lysosomal pathway regulates adult NSC maintenance, the activation of quiescent NSCs, the survival of the newly born neurons and the timing of their maturation. Future research is warranted to validate the results of these pioneering studies, refine the molecular mechanisms underlying the regulation of NSCs and newborn neurons by autophagy throughout the life-span of mammals and provide significance to the autophagic process in adult neurogenesis-dependent behavioral tasks, in physiological and pathological conditions. These lines of research may have important consequences for our understanding of stem cell dysfunction and neurogenic decline during healthy aging and neurodegeneration.
Collapse
Affiliation(s)
- Lucía Casares-Crespo
- Stem Cells and Aging Unit, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, València, Spain
| | - Isabel Calatayud-Baselga
- Stem Cells and Aging Unit, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, València, Spain
| | - Laura García-Corzo
- Stem Cells and Aging Unit, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, València, Spain
| | - Helena Mira
- Stem Cells and Aging Unit, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, València, Spain
| |
Collapse
|
21
|
Ferrer I. Oligodendrogliopathy in neurodegenerative diseases with abnormal protein aggregates: The forgotten partner. Prog Neurobiol 2018; 169:24-54. [DOI: 10.1016/j.pneurobio.2018.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 07/24/2018] [Accepted: 07/27/2018] [Indexed: 12/31/2022]
|
22
|
Yan X, Uronen RL, Huttunen HJ. The interaction of α-synuclein and Tau: A molecular conspiracy in neurodegeneration? Semin Cell Dev Biol 2018; 99:55-64. [PMID: 29738880 DOI: 10.1016/j.semcdb.2018.05.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 02/06/2018] [Accepted: 05/04/2018] [Indexed: 12/18/2022]
Abstract
α-synuclein and Tau are proteins prone to pathological misfolding and aggregation that are normally found in the presynaptic and axonal compartments of neurons. Misfolding initiates a homo-oligomerization and aggregation cascade culminating in cerebral accumulation of aggregated α-synuclein and Tau in insoluble protein inclusions in multiple neurodegenerative diseases. Traditionally, α-synuclein-containing Lewy bodies have been associated with Parkinson's disease and Tau-containing neurofibrillary tangles with Alzheimer's disease and various frontotemporal dementia syndromes. However, there is significant overlap and co-occurrence of α-synuclein and Tau pathologies in a spectrum of neurodegenerative diseases. Importantly, α-synuclein and Tau can interact in cells, and their pathological conformations are capable of templating further misfolding and aggregation of each other. They also share a number of protein interactors indicating that network perturbations may contribute to chronic proteotoxic stress and neuronal dysfunction in synucleinopathies and tauopathies, some of which share similarities in both neuropathological and clinical manifestations. In this review, we focus on the protein interactions of these two pathologically important proteins and consider a network biology perspective towards neurodegenerative diseases.
Collapse
Affiliation(s)
- Xu Yan
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland
| | - Riikka-Liisa Uronen
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland
| | - Henri J Huttunen
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, 00014 Helsinki, Finland.
| |
Collapse
|
23
|
Sen T, Saha P, Sen N. Nitrosylation of GAPDH augments pathological tau acetylation upon exposure to amyloid-β. Sci Signal 2018; 11:11/522/eaao6765. [PMID: 29559585 DOI: 10.1126/scisignal.aao6765] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Acetylation of the microtubule-associated protein tau promotes its polymerization into neurofibrillary tangles that are implicated in the pathology of Alzheimer's disease (AD). The gaseous neurotransmitter nitric oxide (NO) regulates cell signaling through the nitrosylation of proteins. We found that NO production and tau acetylation at Lys280 occurred in the brain tissue in mice and in cultured mouse cortical neurons in response to exposure to amyloid-β1-42 (Aβ1-42), a peptide that is also implicated in AD. An increased abundance of NO facilitated the S-nitrosylation (SNO) of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). S-nitrosylated GAPDH (GAPDH-SNO) promoted the acetylation and activation of the acetyltransferase p300 and facilitated the nitrosylation and inactivation of the deacetylase sirtuin 1 (SIRT1). The abundance of GAPDH-SNO was increased in postmortem brain samples from AD patients. Preventing the increase in GAPDH-SNO abundance in both cultured neurons and mice, either by overexpression of the nitrosylation mutant of GAPDH (GAPDH C150S) or by treatment with the GAPDH nitrosylation inhibitor CGP3466B (also known as omigapil), abrogated Aβ1-42-induced tau acetylation, memory impairment, and locomotor dysfunction in mice, suggesting that this drug might be repurposed to treat patients with AD.
Collapse
Affiliation(s)
- Tanusree Sen
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh, PA 15213, USA
| | - Pampa Saha
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh, PA 15213, USA
| | - Nilkantha Sen
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh, PA 15213, USA.
| |
Collapse
|
24
|
Young ZT, Mok SA, Gestwicki JE. Therapeutic Strategies for Restoring Tau Homeostasis. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a024612. [PMID: 28159830 DOI: 10.1101/cshperspect.a024612] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Normal tau homeostasis is achieved when the synthesis, processing, and degradation of the protein is balanced. Together, the pathways that regulate tau homeostasis ensure that the protein is at the proper levels and that its posttranslational modifications and subcellular localization are appropriately controlled. These pathways include the enzymes responsible for posttranslational modifications, those systems that regulate mRNA splicing, and the molecular chaperones that control tau turnover and its binding to microtubules. In tauopathies, this delicate balance is disturbed. Tau becomes abnormally modified by posttranslational modification, it loses affinity for microtubules, and it accumulates in proteotoxic aggregates. How and why does this imbalance occur? In this review, we discuss how molecular chaperones and other components of the protein homeostasis (e.g., proteostasis) network normally govern tau quality control. We also discuss how aging might reduce the capacity of these systems and how tau mutations might further affect this balance. Finally, we discuss how small-molecule inhibitors are being used to probe and perturb the tau quality-control systems, playing a particularly prominent role in revealing the logic of tau homeostasis. As such, there is now interest in developing these chemical probes into therapeutics, with the goal of restoring normal tau homeostasis to treat disease.
Collapse
Affiliation(s)
- Zapporah T Young
- Institute for Neurodegenerative Disease, Department of Pharmaceutical Chemistry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| | - Sue Ann Mok
- Institute for Neurodegenerative Disease, Department of Pharmaceutical Chemistry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| | - Jason E Gestwicki
- Institute for Neurodegenerative Disease, Department of Pharmaceutical Chemistry, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
25
|
HDAC6 deficiency induces apoptosis in mesenchymal stem cells through p53 K120 acetylation. Biochem Biophys Res Commun 2017; 494:51-56. [DOI: 10.1016/j.bbrc.2017.10.087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/16/2017] [Indexed: 01/05/2023]
|
26
|
The HDAC inhibitor AR42 interacts with pazopanib to kill trametinib/dabrafenib-resistant melanoma cells in vitro and in vivo. Oncotarget 2017; 8:16367-16386. [PMID: 28146421 PMCID: PMC5369969 DOI: 10.18632/oncotarget.14829] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/19/2017] [Indexed: 12/11/2022] Open
Abstract
Studies focused on the killing of activated B-RAF melanoma cells by the histone deacetylase (HDAC) inhibitor AR42. Compared to other tumor cell lines, PDX melanoma isolates were significantly more sensitive to AR42-induced killing. AR42 and the multi-kinase inhibitor pazopanib interacted to activate: an eIF2α–Beclin1 pathway causing autophagosome formation; an eIF2α–DR4/DR5/CD95 pathway; and an eIF2α-dependent reduction in the expression of c-FLIP-s, MCL-1 and BCL-XL. AR42 did not alter basal chaperone activity but increased the ability of pazopanib to inhibit HSP90, HSP70 and GRP78. AR42 and pazopanib caused HSP90/HSP70 dissociation from RAF-1 and B-RAF that resulted in reduced ‘RAF’ expression. The drug combination activated a DNA-damage-ATM-AMPK pathway that was associated with: NFκB activation; reduced mTOR S2448 and ULK-1 S757 phosphorylation; and increased ULK-1 S317 and ATG13 S318 phosphorylation. Knock down of PERK, eIF2α, Beclin1, ATG5 or AMPKα, or expression of IκB S32A S36A, ca-mTOR or TRX, reduced cell killing. AR42, via lysosomal degradation, reduced the protein expression of HDACs 2/5/6/10/11. In vivo, a 3-day exposure of dabrafenib/trametinib resistant melanoma cells to the AR42 pazopanib combination reduced tumor growth and enhanced survival from ∼25 to ∼40 days. Tumor cells that had adapted through therapy exhibited elevated HGF expression and the c-MET inhibitor crizotinib enhanced AR42 pazopanib lethality in this evolved drug-resistant population.
Collapse
|
27
|
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
|
28
|
Šimić G, Babić Leko M, Wray S, Harrington CR, Delalle I, Jovanov-Milošević N, Bažadona D, Buée L, de Silva R, Di Giovanni G, Wischik CM, Hof PR. Monoaminergic neuropathology in Alzheimer's disease. Prog Neurobiol 2017; 151:101-138. [PMID: 27084356 PMCID: PMC5061605 DOI: 10.1016/j.pneurobio.2016.04.001] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/09/2016] [Accepted: 04/05/2016] [Indexed: 01/02/2023]
Abstract
None of the proposed mechanisms of Alzheimer's disease (AD) fully explains the distribution patterns of the neuropathological changes at the cellular and regional levels, and their clinical correlates. One aspect of this problem lies in the complex genetic, epigenetic, and environmental landscape of AD: early-onset AD is often familial with autosomal dominant inheritance, while the vast majority of AD cases are late-onset, with the ε4 variant of the gene encoding apolipoprotein E (APOE) known to confer a 5-20 fold increased risk with partial penetrance. Mechanisms by which genetic variants and environmental factors influence the development of AD pathological changes, especially neurofibrillary degeneration, are not yet known. Here we review current knowledge of the involvement of the monoaminergic systems in AD. The changes in the serotonergic, noradrenergic, dopaminergic, histaminergic, and melatonergic systems in AD are briefly described. We also summarize the possibilities for monoamine-based treatment in AD. Besides neuropathologic AD criteria that include the noradrenergic locus coeruleus (LC), special emphasis is given to the serotonergic dorsal raphe nucleus (DRN). Both of these brainstem nuclei are among the first to be affected by tau protein abnormalities in the course of sporadic AD, causing behavioral and cognitive symptoms of variable severity. The possibility that most of the tangle-bearing neurons of the LC and DRN may release amyloid β as well as soluble monomeric or oligomeric tau protein trans-synaptically by their diffuse projections to the cerebral cortex emphasizes their selective vulnerability and warrants further investigations of the monoaminergic systems in AD.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Selina Wray
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | | | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Nataša Jovanov-Milošević
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Danira Bažadona
- Department of Neurology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Luc Buée
- University of Lille, Inserm, CHU-Lille, UMR-S 1172, Alzheimer & Tauopathies, Lille, France
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Claude M Wischik
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Patrick R Hof
- Fishberg Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
29
|
Shi Y, Xu L, Tang J, Fang L, Ma S, Ma X, Nie J, Pi X, Qiu A, Zhuang S, Liu N. Inhibition of HDAC6 protects against rhabdomyolysis-induced acute kidney injury. Am J Physiol Renal Physiol 2017; 312:F502-F515. [PMID: 28052874 DOI: 10.1152/ajprenal.00546.2016] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/29/2016] [Accepted: 12/29/2016] [Indexed: 12/18/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) inhibition has been reported to protect against ischemic stroke and prolong survival after sepsis in animal models. However, it remains unknown whether HDAC6 inhibition offers a renoprotective effect after acute kidney injury (AKI). In this study, we examined the effect of tubastatin A (TA), a highly selective inhibitor of HDAC6, on AKI in a murine model of glycerol (GL) injection-induced rhabdomyolysis. Following GL injection, the mice developed severe acute tubular injury as indicated by renal dysfunction; expression of neutrophil gelatinase-associated lipocalin (NGAL), an injury marker of renal tubules; and an increase of TdT-mediated dUTP nick-end labeling (TUNEL)-positive tubular cells. These changes were companied by increased HDAC6 expression in the cytoplasm of renal tubular cells. Administration of TA significantly reduced serum creatinine and blood urea nitrogen levels as well as attenuated renal tubular damage in injured kidneys. HDAC6 inhibition also resulted in decreased expression of NGAL, reduced apoptotic cell, and inactivated caspase-3 in the kidney after acute injury. Moreover, injury to the kidney increased phosphorylation of nuclear factor (NF)-κB and expression of multiple cytokines/chemokines including tumor necrotic factor-α and interleukin-6 and monocyte chemoattractant protein-1, as well as macrophage infiltration. Treatment with TA attenuated all those responses. Finally, HDAC6 inhibition reduced the level of oxidative stress by suppressing malondialdehyde (MDA) and preserving expression of superoxide dismutase (SOD) in the injured kidney. Collectively, these data indicate that HDAC6 contributes to the pathogenesis of rhabdomyolysis-induced AKI and suggest that HDAC6 inhibitors have therapeutic potential for AKI treatment.
Collapse
Affiliation(s)
- Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liuqing Xu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinhua Tang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu Fang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuchen Ma
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyan Ma
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Nie
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoling Pi
- Department of Internal Medicine, Pudong New District Gongli Hospital, Shanghai, China
| | - Andong Qiu
- School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai, China; and
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Brown University School of Medicine, Providence, Rhode Island
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China;
| |
Collapse
|
30
|
Kumar V, Sami N, Kashav T, Islam A, Ahmad F, Hassan MI. Protein aggregation and neurodegenerative diseases: From theory to therapy. Eur J Med Chem 2016; 124:1105-1120. [DOI: 10.1016/j.ejmech.2016.07.054] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 07/20/2016] [Accepted: 07/22/2016] [Indexed: 12/23/2022]
|
31
|
Tyson T, Steiner JA, Brundin P. Sorting out release, uptake and processing of alpha-synuclein during prion-like spread of pathology. J Neurochem 2016; 139 Suppl 1:275-289. [PMID: 26617280 PMCID: PMC4958606 DOI: 10.1111/jnc.13449] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 12/17/2022]
Abstract
Parkinson's disease is a progressive neurological disorder that is characterized by the formation of intracellular protein inclusion bodies composed primarily of a misfolded and aggregated form of the protein α-synuclein. There is growing evidence that supports the prion-like hypothesis of α-synuclein progression. This hypothesis postulates that α-synuclein is a prion-like pathological agent and is responsible for the progression of Parkinson pathology in the brain. Potential misfolding or aggregation of α-synuclein that might occur in the peripheral nervous system as a result of some insult, environmental or genetic (or more likely a combination of both) that might spread into the midbrain, eventually causing degeneration of the neurons in the substantia nigra. As the disease progresses further, it is likely that α-synuclein pathology continues to spread throughout the brain, including the cortex, leading to deterioration of cognition and higher brain functions. While it is unknown why α-synuclein initially misfolds and aggregates, a great deal has been learned about how the cell handles aberrant α-synuclein assemblies. In this review, we focus on these mechanisms and discuss them in an attempt to define the role that they might play in the propagation of misfolded α-synuclein from cell-to-cell. The prion-like hypothesis of α-synuclein pathology suggests a method for the transmission of misfolded α-synuclein from one neuron to another. This hypothesis postulates that misfolded α-synuclein becomes aggregation prone and when released and taken up by neighboring cells, seeds further misfolding and aggregation. In this review we examine the cellular mechanisms that are involved in the processing of α-synuclein and how these may contribute to the prion-like propagation of α-synuclein pathology. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Trevor Tyson
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Jennifer A Steiner
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA.
| |
Collapse
|
32
|
Cecarini V, Bonfili L, Cuccioloni M, Mozzicafreddo M, Angeletti M, Keller JN, Eleuteri AM. The fine-tuning of proteolytic pathways in Alzheimer's disease. Cell Mol Life Sci 2016; 73:3433-51. [PMID: 27120560 PMCID: PMC11108445 DOI: 10.1007/s00018-016-2238-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/31/2016] [Accepted: 04/21/2016] [Indexed: 11/28/2022]
Abstract
Several integrated proteolytic systems contribute to the maintenance of cellular homeostasis through the continuous removal of misfolded, aggregated or oxidized proteins and damaged organelles. Among these systems, the proteasome and autophagy play the major role in protein quality control, which is a fundamental issue in non-proliferative cells such as neurons. Disturbances in the functionality of these two pathways are frequently observed in neurodegenerative diseases, like Alzheimer's disease, and reflect the accumulation of protease-resistant, deleterious protein aggregates. In this review, we explored the sophisticated crosstalk between the ubiquitin-proteasome system and autophagy in the removal of the harmful structures that characterize Alzheimer's disease neurons. We also dissected the role of the numerous shuttle factors and chaperones that, directly or indirectly interacting with ubiquitin and LC3, are used for cargo selection and delivery to one pathway or the other.
Collapse
Affiliation(s)
- Valentina Cecarini
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy.
| | - Laura Bonfili
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| | - Massimiliano Cuccioloni
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| | - Matteo Mozzicafreddo
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| | - Mauro Angeletti
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| | - Jeffrey N Keller
- Pennington Biomedical Research Centre, Louisiana State University System, Baton Rouge, LA, 70808, USA
| | - Anna Maria Eleuteri
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| |
Collapse
|
33
|
Arendt T, Stieler JT, Holzer M. Tau and tauopathies. Brain Res Bull 2016; 126:238-292. [PMID: 27615390 DOI: 10.1016/j.brainresbull.2016.08.018] [Citation(s) in RCA: 424] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 12/11/2022]
|
34
|
Fan T, Huang Z, Chen L, Wang W, Zhang B, Xu Y, Pan S, Mao Z, Hu H, Geng Q. Associations between autophagy, the ubiquitin-proteasome system and endoplasmic reticulum stress in hypoxia-deoxygenation or ischemia-reperfusion. Eur J Pharmacol 2016; 791:157-167. [PMID: 27568838 DOI: 10.1016/j.ejphar.2016.08.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 11/25/2022]
Abstract
The activation of autophagy has been demonstrated to exert protective roles during hypoxia-reoxygenation (H/R)-induced brain injuries. This study aimed to investigate whether and how preconditioning with a proteasome inhibitor (MG-132), a proteasome promoter (Adriamycin, ADM), an autophagy inhibitor (3-methyladenine, 3-MA) and an autophagy promoter (Rapamycin, Rap) affected endoplasmic reticulum stress (ERS), the ubiquitin-proteasome system (UPS), autophagy, inflammation and apoptosis. Ubiquitin protein and 26S proteasome activity levels were decreased by MG-132 pretreatment but increased by ADM pretreatment at 2h, 4h and 6h following H/R treatment. MG-132 pretreatment led to the increased expression of autophagy-related genes, ER stress-associated genes and IκB but decreased the expression levels of NF-κB and caspase-3. ADM pretreatment led to the decreased expression of autophagy-related genes, ERS-associated genes and IκB but increased the expression of NF-κB and caspase-3. Pretreatment with 3-MA reduced the expression of autophagy-related genes, autophagy and UPS co-related genes, as well as apoptosis-related although the latter was increased by Rap pretreatment at 2h, 4h and 6h following H/R treatment. In vivo, pretreatment of rats with ADM, MG-132, 3-MA or Rap followed by ischemia-reperfusion (I/R) treatment resulted in similar changes. Proteasome inhibition preconditioning strengthened autophagy and ER stress but decreased apoptosis and inflammation. Autophagy promotion preconditioning exhibited similar changes. The combination of a proteasome inhibitor and an autophagy promoter might represent a new possible therapy to treat H/R or I/R injury-related diseases.
Collapse
Affiliation(s)
- Tao Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhixin Huang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Chen
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Boyou Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yao Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shize Pan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhangfan Mao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Hu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
35
|
Boman A, Svensson S, Boxer A, Rojas JC, Seeley WW, Karydas A, Miller B, Kågedal K, Svenningsson P. Distinct Lysosomal Network Protein Profiles in Parkinsonian Syndrome Cerebrospinal Fluid. JOURNAL OF PARKINSON'S DISEASE 2016; 6:307-15. [PMID: 27061067 PMCID: PMC4927933 DOI: 10.3233/jpd-150759] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/13/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND Clinical diagnosis of parkinsonian syndromes like Parkinson's disease (PD), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP) is hampered by overlapping symptomatology and lack of diagnostic biomarkers, and definitive diagnosis is only possible post-mortem. OBJECTIVE Since impaired protein degradation plays an important role in many neurodegenerative disorders, we hypothesized that profiles of select lysosomal network proteins in cerebrospinal fluid could be differentially expressed in these parkinsonian syndromes. METHODS Cerebrospinal fluid samples were collected from PD patients (n = 18), clinically diagnosed 4-repeat tauopathy patients; corticobasal syndrome (CBS) (n = 3) and PSP (n = 8); and pathologically diagnosed PSP (n = 8) and CBD patients (n = 7). Each patient set was compared to its appropriate control group consisting of age and gender matched individuals. Select lysosomal network protein levels were detected via Western blotting. Factor analysis was used to test the diagnostic sensitivity, specificity and accuracy of the select lysosomal network protein expression profiles. RESULTS PD, CBD and PSP were markedly different in their cerebrospinal fluid lysosomal network protein profiles. Lysosomal-associated membrane proteins 1 and 2 were significantly decreased in PD; early endosomal antigen 1 was decreased and lysozyme increased in PSP; and lysosomal-associated membrane proteins 1 and 2, microtubule-associated protein 1 light chain 3 and lysozyme were increased in CBD. A panel of lysosomal-associated membrane protein 2, lysozyme and microtubule-associated protein 1 light chain discriminated between controls, PD and 4-repeat tauopathies. CONCLUSIONS This study offers proof of concept that select lysosomal network proteins are differentially expressed in cerebrospinal fluid of Parkinson's disease, corticobasal syndrome and progressive supranuclear palsy. Lysosomal network protein analysis could be further developed as a diagnostic fluid biomarker in parkinsonian syndromes.
Collapse
Affiliation(s)
- Andrea Boman
- Experimental Pathology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
- Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Samuel Svensson
- Developmental Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- CBD Solutions, Stockholm, Sweden
| | - Adam Boxer
- Memory and Aging Center, University of California, San Francisco, USA
| | - Julio C. Rojas
- Memory and Aging Center, University of California, San Francisco, USA
| | - William W. Seeley
- Memory and Aging Center, University of California, San Francisco, USA
| | - Anna Karydas
- Memory and Aging Center, University of California, San Francisco, USA
| | - Bruce Miller
- Memory and Aging Center, University of California, San Francisco, USA
| | - Katarina Kågedal
- Experimental Pathology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Per Svenningsson
- Translational Neuropharmacology, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
36
|
Piras A, Collin L, Grüninger F, Graff C, Rönnbäck A. Autophagic and lysosomal defects in human tauopathies: analysis of post-mortem brain from patients with familial Alzheimer disease, corticobasal degeneration and progressive supranuclear palsy. Acta Neuropathol Commun 2016; 4:22. [PMID: 26936765 PMCID: PMC4774096 DOI: 10.1186/s40478-016-0292-9] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 01/01/2023] Open
Abstract
Introduction The accumulation of insoluble proteins within neurons and glia cells is a pathological hallmark of several neurodegenerative diseases. Abnormal aggregation of the microtubule-associated protein tau characterizes the neuropathology of tauopathies, such as Alzheimer disease (AD), corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP). An impairment of the lysosomal degradation pathway called macroautophagy, hereafter referred to as autophagy, could contribute to the accumulation of aggregated proteins. The role of autophagy in neurodegeneration has been intensively studied in the context of AD but there are few studies in other tauopathies and it is not known if defects in autophagy is a general feature of tauopathies. In the present study, we analysed autophagic and lysosomal markers in human post-mortem brain samples from patients with early-onset familial AD (FAD) with the APP Swedish mutation (APPswe), CBD and PSP and control individuals. Results FAD, CBD and PSP patients displayed an increase in LC3-positive vesicles in frontal cortex, indicating an accumulation of autophagic vesicles. Moreover, using double-immunohistochemistry and in situ proximity ligation assay, we observed colocalization of hyperphosphorylated tau with the autophagy marker LC3 in FAD, CBD and PSP patients but not in control individuals. Increased levels of the lysosomal marker LAMP1 was detected in FAD and CBD, and in addition Cathepsin D was diffusely spread in the cytoplasm in all tauopathies suggesting an impaired lysosomal integrity. Conclusion Taken together, our results indicate an accumulation of autophagic and lysosomal markers in human brain tissue from patients with primary tauopathies (CBD and PSP) as well as FAD, suggesting a defect of the autophagosome-lysosome pathway that may contribute to the development of tau pathology. Electronic supplementary material The online version of this article (doi:10.1186/s40478-016-0292-9) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
Kahlson MA, Colodner KJ. Glial Tau Pathology in Tauopathies: Functional Consequences. J Exp Neurosci 2016; 9:43-50. [PMID: 26884683 PMCID: PMC4750898 DOI: 10.4137/jen.s25515] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 12/22/2022] Open
Abstract
Tauopathies are a class of neurodegenerative diseases characterized by the presence of hyperphosphorylated and aggregated tau pathology in neuronal and glial cells. Though the ratio of neuronal and glial tau aggregates varies across diseases, glial tau aggregates can populate the same degenerating brain regions as neuronal tau aggregates. While much is known about the deleterious consequences of tau pathology in neurons, the relative contribution of glial tau pathology to these diseases is less clear. Recent studies using a number of model systems implicate glial tau pathology in contributing to tauopathy pathogenesis. This review aims to highlight the functional consequences of tau overexpression in glial cells and explore the potential contribution of glial tau pathology in the pathogenesis of neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Martha A Kahlson
- Department of Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, USA
| | - Kenneth J Colodner
- Department of Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, USA
| |
Collapse
|
38
|
Šimić G, Babić Leko M, Wray S, Harrington C, Delalle I, Jovanov-Milošević N, Bažadona D, Buée L, de Silva R, Di Giovanni G, Wischik C, Hof PR. Tau Protein Hyperphosphorylation and Aggregation in Alzheimer's Disease and Other Tauopathies, and Possible Neuroprotective Strategies. Biomolecules 2016; 6:6. [PMID: 26751493 PMCID: PMC4808800 DOI: 10.3390/biom6010006] [Citation(s) in RCA: 458] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 11/28/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022] Open
Abstract
Abnormal deposition of misprocessed and aggregated proteins is a common final pathway of most neurodegenerative diseases, including Alzheimer's disease (AD). AD is characterized by the extraneuronal deposition of the amyloid β (Aβ) protein in the form of plaques and the intraneuronal aggregation of the microtubule-associated protein tau in the form of filaments. Based on the biochemically diverse range of pathological tau proteins, a number of approaches have been proposed to develop new potential therapeutics. Here we discuss some of the most promising ones: inhibition of tau phosphorylation, proteolysis and aggregation, promotion of intra- and extracellular tau clearance, and stabilization of microtubules. We also emphasize the need to achieve a full understanding of the biological roles and post-translational modifications of normal tau, as well as the molecular events responsible for selective neuronal vulnerability to tau pathology and its propagation. It is concluded that answering key questions on the relationship between Aβ and tau pathology should lead to a better understanding of the nature of secondary tauopathies, especially AD, and open new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia.
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia.
| | - Selina Wray
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK.
| | - Charles Harrington
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston 02118, MA, USA.
| | - Nataša Jovanov-Milošević
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia.
| | - Danira Bažadona
- Department of Neurology, University Hospital Center Zagreb, Zagreb 10000, Croatia.
| | - Luc Buée
- Laboratory Alzheimer & Tauopathies, Université Lille and INSERM U1172, Jean-Pierre Aubert Research Centre, Lille 59045, France.
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK.
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta.
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK.
| | - Claude Wischik
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| | - Patrick R Hof
- Fishberg Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
39
|
The Autotaxin-Lysophosphatidic Acid Axis Modulates Histone Acetylation and Gene Expression during Oligodendrocyte Differentiation. J Neurosci 2015; 35:11399-414. [PMID: 26269646 DOI: 10.1523/jneurosci.0345-15.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED During development, oligodendrocytes (OLGs), the myelinating cells of the CNS, undergo a stepwise progression during which OLG progenitors, specified from neural stem/progenitor cells, differentiate into fully mature myelinating OLGs. This progression along the OLG lineage is characterized by well synchronized changes in morphology and gene expression patterns. The latter have been found to be particularly critical during the early stages of the lineage, and they have been well described to be regulated by epigenetic mechanisms, especially by the activity of the histone deacetylases HDAC1 and HDAC2. The data presented here identify the extracellular factor autotaxin (ATX) as a novel upstream signal modulating HDAC1/2 activity and gene expression in cells of the OLG lineage. Using the zebrafish as an in vivo model system as well as rodent primary OLG cultures, this functional property of ATX was found to be mediated by its lysophospholipase D (lysoPLD) activity, which has been well characterized to generate the lipid signaling molecule lysophosphatidic acid (LPA). More specifically, the lysoPLD activity of ATX was found to modulate HDAC1/2 regulated gene expression during a time window coinciding with the transition from OLG progenitor to early differentiating OLG. In contrast, HDAC1/2 regulated gene expression during the transition from neural stem/progenitor to OLG progenitor appeared unaffected by ATX and its lysoPLD activity. Thus, together, our data suggest that an ATX-LPA-HDAC1/2 axis regulates OLG differentiation specifically during the transition from OLG progenitor to early differentiating OLG and via a molecular mechanism that is evolutionarily conserved from at least zebrafish to rodent. SIGNIFICANCE STATEMENT The formation of the axon insulating and supporting myelin sheath by differentiating oligodendrocytes (OLGs) in the CNS is considered an essential step during vertebrate development. In addition, loss and/or dysfunction of the myelin sheath has been associated with a variety of neurologic diseases in which repair is limited, despite the presence of progenitor cells with the potential to differentiate into myelinating OLGs. This study characterizes the autotaxin-lysophosphatidic acid signaling axis as a modulator of OLG differentiation in vivo in the developing zebrafish and in vitro in rodent OLGs in culture. These findings provide novel insight into the regulation of developmental myelination, and they are likely to lead to advancing studies related to the stimulation of myelin repair under pathologic conditions.
Collapse
|
40
|
Lopes da Fonseca T, Villar-Piqué A, Outeiro TF. The Interplay between Alpha-Synuclein Clearance and Spreading. Biomolecules 2015; 5:435-71. [PMID: 25874605 PMCID: PMC4496680 DOI: 10.3390/biom5020435] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 12/23/2022] Open
Abstract
Parkinson's Disease (PD) is a complex neurodegenerative disorder classically characterized by movement impairment. Pathologically, the most striking features of PD are the loss of dopaminergic neurons and the presence of intraneuronal protein inclusions primarily composed of alpha-synuclein (α-syn) that are known as Lewy bodies and Lewy neurites in surviving neurons. Though the mechanisms underlying the progression of PD pathology are unclear, accumulating evidence suggests a prion-like spreading of α-syn pathology. The intracellular homeostasis of α-syn requires the proper degradation of the protein by three mechanisms: chaperone-mediated autophagy, macroautophagy and ubiquitin-proteasome. Impairment of these pathways might drive the system towards an alternative clearance mechanism that could involve its release from the cell. This increased release to the extracellular space could be the basis for α-syn propagation to different brain areas and, ultimately, for the spreading of pathology and disease progression. Here, we review the interplay between α-syn degradation pathways and its intercellular spreading. The understanding of this interplay is indispensable for obtaining a better knowledge of the molecular basis of PD and, consequently, for the design of novel avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Tomás Lopes da Fonseca
- Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen 37073, Germany.
- Instituto de Fisiologia, Faculty of Medicine, University of Lisbon, Lisboa 1649-028, Portugal.
| | - Anna Villar-Piqué
- Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen 37073, Germany.
| | - Tiago Fleming Outeiro
- Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen 37073, Germany.
- Instituto de Fisiologia, Faculty of Medicine, University of Lisbon, Lisboa 1649-028, Portugal.
- CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa 1150, Portugal.
| |
Collapse
|