1
|
Samson JS, Rajagopal K, Parvathi VD. Outlook of SNCA (α-synuclein) transgenic fly models in delineating the sequel of mitochondrial dysfunction in Parkinson's disease. Brain Res 2025; 1852:149505. [PMID: 39954798 DOI: 10.1016/j.brainres.2025.149505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/21/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with mechanisms that results in loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the brain. Being a complex heterogeneous disorder, there is a requisite in discovering the underlying molecular signatures that could potentially help in resolving challenges associated with diagnosis as well as therapeutic management. SNCA gene that encodes for the protein α-synuclein is widely known for its indispensable role in aggravating the progression of sporadic and familial PD, upon mutations. Likewise, mitochondrial dysfunction is inferred to be playing a central role in both forms of PD. Observations from experimental models and human PD cases displayed strong evidence for disruption of mitochondrial dynamics, inhibition of mitochondrial complex I protein's function and elevation in reactive oxygen species (ROS) by the toxic aggregation of α-synuclein. Further, recent studies have raised the possibility of an underlying relationship, where the α-synuclein toxicity is exacerbated by the mutant mitochondrial complex proteins and vice-versa. In this review, we provide an overview of mechanisms influencing α-synuclein-related neurodegeneration, particularly, emphasizing the role of SNCA (α-synuclein) gene in leading to altered mitochondrial biogenesis during PD. We have described how transgenic Drosophila models were reliable at recapitulating some of the essential characteristics of PD. In addition, we highlight the capability of utilizing transgenic fly models in deciphering the altered α-synuclein toxicity and mitochondrial dysfunction, as induced by defects in the mitochondrial DNA.
Collapse
Affiliation(s)
- Jennifer Sally Samson
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India.
| | | | - Venkatachalam Deepa Parvathi
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India.
| |
Collapse
|
2
|
Li X, Wang W, Pan S, Cao X, Thomas ER, Xie M, Zhang C, Wu J. Exploring heat shock proteins as therapeutic targets for Parkinson's disease. Biochem Pharmacol 2024; 230:116633. [PMID: 39551273 DOI: 10.1016/j.bcp.2024.116633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
Parkinson's disease (PD) is characterized by the accumulation of misfolded α-synuclein (α-syn). Promoting the degradation of misfolded proteins has been shown to be an effective approach to alleviate PD. This review highlights the roles of specific heat shock proteins (HSPs) in modulating α-syn aggregation and neuronal survival. HSP27 prevents glycosylation-induced α-syn aggregation, disrupts copper ion interactions, inhibits mitochondrial apoptosis, and prevents dopaminergic neuronal cell death. HSP70 alleviates dopaminergic neuronal damage by promoting mitophagy and preventing neuronal apoptosis. HSC70 plays a critical role in chaperone-mediated autophagy and facilitates lysosomal degradation. GRP78 mitigates abnormal protein aggregation. The HSP70-HSP40-HSP110 system is capable of degrading α-syn amyloid fibers. Inhibition of HSP90 expression protects neurons. Further research should prioritize developing regulators of HSPs as treatments for PD. While HSPs offer promise in PD management, their complex roles necessitate cautious therapeutic development to harness their potential. Understanding the specific roles of different HSPs will be essential to developing effective therapies for α-syn clearance.
Collapse
Affiliation(s)
- Xiang Li
- The Zigong Affiliated Hospital, Southwest Medical University, Zigong Mental Health Center, Zigong Institute of Brain Science, Zigong, Sichuan Province 643020, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Wenjun Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Shi Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Xueqin Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | | | - Mingyu Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Chunxiang Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China.
| | - Jianming Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
3
|
Uversky VN. How to drug a cloud? Targeting intrinsically disordered proteins. Pharmacol Rev 2024; 77:PHARMREV-AR-2023-001113. [PMID: 39433443 DOI: 10.1124/pharmrev.124.001113] [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: 06/16/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024] Open
Abstract
Biologically active proteins/regions without stable structure (i.e., intrinsically disordered proteins and regions (IDPs and IDRs)) are commonly found in all proteomes. They have a unique functional repertoire that complements the functionalities of ordered proteins and domains. IDPs/IDRs are multifunctional promiscuous binders capable of folding at interaction with specific binding partners on a template- or context-dependent manner, many of which undergo liquid-liquid phase separation, leading to the formation of membrane-less organelles and biomolecular condensates. Many of them are frequently related to the pathogenesis of various human diseases. All this defines IDPs/IDRs as attractive targets for the development of novel drugs. However, their lack of unique structures, multifunctionality, binding promiscuity, and involvement in unusual modes of action preclude direct use of traditional structure-based drug design approaches for targeting IDPs/IDRs, and make disorder-based drug discovery for these "protein clouds" challenging. Despite all these complexities there is continuing progress in the design of small molecules affecting IDPs/IDRs. This article describes the major structural features of IDPs/IDRs and the peculiarities of the disorder-based functionality. It also discusses the roles of IDPs/IDRs in various pathologies, and shows why the approaches elaborated for finding drugs targeting ordered proteins cannot be directly used for the intrinsic disorder-based drug design, and introduces some novel methodologies suitable for these purposes. Finally, it emphasizes that regardless of their multifunctionality, binding promiscuity, lack of unique structures, and highly dynamic nature, "protein clouds" are principally druggable. Significance Statement Intrinsically disordered proteins and regions are highly abundant in nature, have multiple important biological functions, are commonly involved in the pathogenesis of a multitude of human diseases, and are therefore considered as very attractive drug targets. Although dealing with these unstructured multifunctional protein/regions is a challenging task, multiple innovative approaches have been designed to target them by small molecules.
Collapse
|
4
|
Mohammed S, Russo I, Ramazzina I. Uncovering the Role of Natural and Synthetic Small Molecules in Counteracting the Burden of α-Synuclein Aggregates and Related Toxicity in Different Models of Parkinson's Disease. Int J Mol Sci 2023; 24:13370. [PMID: 37686175 PMCID: PMC10488152 DOI: 10.3390/ijms241713370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
A proteostasis network represents a sophisticated cellular system that controls the whole process which leads to properly folded functional proteins. The imbalance of proteostasis determines a quantitative increase in misfolded proteins prone to aggregation and elicits the onset of different diseases. Among these, Parkinson's Disease (PD) is a progressive brain disorder characterized by motor and non-motor signs. In PD pathogenesis, alpha-Synuclein (α-Syn) loses its native structure, triggering a polymerization cascade that leads to the formation of toxic inclusions, the PD hallmark. Because molecular chaperones represent a "cellular arsenal" to counteract protein misfolding and aggregation, the modulation of their expression represents a compelling PD therapeutic strategy. This review will discuss evidence concerning the effects of natural and synthetic small molecules in counteracting α-Syn aggregation process and related toxicity, in different in vitro and in vivo PD models. Firstly, the role of small molecules that modulate the function(s) of chaperones will be highlighted. Then, attention will be paid to small molecules that interfere with different steps of the protein-aggregation process. This overview would stimulate in-depth research on already-known small molecules or the development of new ones, with the aim of developing drugs that are able to modify the progression of the disease.
Collapse
Affiliation(s)
- Salihu Mohammed
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy;
| | - Isabella Russo
- Department of Molecular and Translational Medicine, University of Brescia, Via Europa 11, 25123 Brescia, Italy;
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni 4, 25125 Brescia, Italy
| | - Ileana Ramazzina
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy;
- Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, 43124 Parma, Italy
- Biostructures and Biosystems National Institute (INBB), Viale Medaglie d’Oro 305, 00136 Rome, Italy
| |
Collapse
|
5
|
Bonavita R, Scerra G, Di Martino R, Nuzzo S, Polishchuk E, Di Gennaro M, Williams SV, Caporaso MG, Caiazza C, Polishchuk R, D’Agostino M, Fleming A, Renna M. The HSPB1-p62/SQSTM1 functional complex regulates the unconventional secretion and transcellular spreading of the HD-associated mutant huntingtin protein. Hum Mol Genet 2023; 32:2269-2291. [PMID: 36971475 PMCID: PMC10321397 DOI: 10.1093/hmg/ddad047] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/06/2023] [Accepted: 03/23/2023] [Indexed: 07/20/2023] Open
Abstract
Conformational diseases, such as Alzheimer, Parkinson and Huntington diseases, are part of a common class of neurological disorders characterized by the aggregation and progressive accumulation of proteins bearing aberrant conformations. Huntington disease (HD) has autosomal dominant inheritance and is caused by mutations leading to an abnormal expansion in the polyglutamine (polyQ) tract of the huntingtin (HTT) protein, leading to the formation of HTT inclusion bodies in neurons of affected patients. Interestingly, recent experimental evidence is challenging the conventional view by which the disease pathogenesis is solely a consequence of the intracellular accumulation of mutant protein aggregates. These studies reveal that transcellular transfer of mutated huntingtin protein is able to seed oligomers involving even the wild-type (WT) forms of the protein. To date, there is still no successful strategy to treat HD. Here, we describe a novel functional role for the HSPB1-p62/SQSTM1 complex, which acts as a cargo loading platform, allowing the unconventional secretion of mutant HTT by extracellular vesicles. HSPB1 interacts preferentially with polyQ-expanded HTT compared with the WT protein and affects its aggregation. Furthermore, HSPB1 levels correlate with the rate of mutant HTT secretion, which is controlled by the activity of the PI3K/AKT/mTOR signalling pathway. Finally, we show that these HTT-containing vesicular structures are biologically active and able to be internalized by recipient cells, therefore providing an additional mechanism to explain the prion-like spreading properties of mutant HTT. These findings might also have implications for the turn-over of other disease-associated, aggregation-prone proteins.
Collapse
Affiliation(s)
| | | | - R Di Martino
- Institute for Endocrinology and Experimental Oncology “G. Salvatore,” National Research Council, 80131 Naples, Italy
- Institute of Biochemistry and Cell Biology, National Research Council, 80131 Naples, Italy
| | - S Nuzzo
- IRCCS SYNLAB SDN, 80143 Naples, Italy
| | - E Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - M Di Gennaro
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy
| | - S V Williams
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - M G Caporaso
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy
| | - C Caiazza
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy
| | - R Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - M D’Agostino
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy
| | - A Fleming
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - M Renna
- To whom correspondence should be addressed at: Department of Molecular Medicine and Medical Biotechnologies, School of Medicine, University of Naples “Federico II”, Via S. Pansini, 5, Building 19, Corpi Bassi Sud (I floor), 80131 Naples, Italy. Tel: +39 081/7463623, Fax: +39 081-7463205;
| |
Collapse
|
6
|
Lee RMQ, Koh TW. Genetic modifiers of synucleinopathies-lessons from experimental models. OXFORD OPEN NEUROSCIENCE 2023; 2:kvad001. [PMID: 38596238 PMCID: PMC10913850 DOI: 10.1093/oons/kvad001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2024]
Abstract
α-Synuclein is a pleiotropic protein underlying a group of progressive neurodegenerative diseases, including Parkinson's disease and dementia with Lewy bodies. Together, these are known as synucleinopathies. Like all neurological diseases, understanding of disease mechanisms is hampered by the lack of access to biopsy tissues, precluding a real-time view of disease progression in the human body. This has driven researchers to devise various experimental models ranging from yeast to flies to human brain organoids, aiming to recapitulate aspects of synucleinopathies. Studies of these models have uncovered numerous genetic modifiers of α-synuclein, most of which are evolutionarily conserved. This review discusses what we have learned about disease mechanisms from these modifiers, and ways in which the study of modifiers have supported ongoing efforts to engineer disease-modifying interventions for synucleinopathies.
Collapse
Affiliation(s)
- Rachel Min Qi Lee
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore, 117604, Singapore
| | - Tong-Wey Koh
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore, 117604, Singapore
- Department of Biological Sciences, National University of Singapore, Block S3 #05-01, 16 Science Drive 4, Singapore, 117558, Singapore
| |
Collapse
|
7
|
Kulkarni A, Preeti K, Tryphena KP, Srivastava S, Singh SB, Khatri DK. Proteostasis in Parkinson's disease: Recent development and possible implication in diagnosis and therapeutics. Ageing Res Rev 2023; 84:101816. [PMID: 36481490 DOI: 10.1016/j.arr.2022.101816] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/27/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
The protein dyshomeostasis is identified as the hallmark of many age-related neurodegenerative disorders including Parkinson's disease (PD). The diseased brain shows the deposition of Lewy bodies composed of α-synuclein protein aggregates. Functional proteostasis is characterized by the well-coordinated signaling network constituting unfolded protein response (UPR), the ubiquitin-proteasome system (UPS), and the autophagy-lysosome pathway (ALP). These networks ensure proper synthesis, folding, confirmation, and degradation of protein i.e., α-synuclein protein in PD. The proper functioning the of intricately woven proteostasis network is quite resilient to sustain under the influence of stressors. The synuclein protein turnover is hugely influenced by the autosomal dominant, recessive, and X-linked mutational changes of a gene involved in UPR, UPS, and ALP. The methylation, acetylation-related epigenetic modifications of DNA and histone proteins along with microRNA-mediated transcriptional changes also lead to extensive proteostasis dysregulation. The result of defective proteostasis is the deposition of many proteins which start appearing in the biofluids and can be identified as potential biomarkers for early diagnosis of PD. The therapeutic intervention targeted at different strata of proteostasis machinery holds great possibilities for delaying the age-related accumulation of pathological hallmarks.
Collapse
Affiliation(s)
- Amrita Kulkarni
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Kumari Preeti
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Kamatham Pushpa Tryphena
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Shashi Bala Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education, and Research (NIPER), Hyderabad, Telangana 500037, India.
| |
Collapse
|
8
|
The Role of Small Heat Shock Proteins in Protein Misfolding Associated Motoneuron Diseases. Int J Mol Sci 2022; 23:ijms231911759. [PMID: 36233058 PMCID: PMC9569637 DOI: 10.3390/ijms231911759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Motoneuron diseases (MNDs) are neurodegenerative conditions associated with death of upper and/or lower motoneurons (MNs). Proteostasis alteration is a pathogenic mechanism involved in many MNDs and is due to the excessive presence of misfolded and aggregated proteins. Protein misfolding may be the product of gene mutations, or due to defects in the translation process, or to stress agents; all these conditions may alter the native conformation of proteins making them prone to aggregate. Alternatively, mutations in members of the protein quality control (PQC) system may determine a loss of function of the proteostasis network. This causes an impairment in the capability to handle and remove aberrant or damaged proteins. The PQC system consists of the degradative pathways, which are the autophagy and the proteasome, and a network of chaperones and co-chaperones. Among these components, Heat Shock Protein 70 represents the main factor in substrate triage to folding, refolding, or degradation, and it is assisted in this task by a subclass of the chaperone network, the small heat shock protein (sHSPs/HSPBs) family. HSPBs take part in proteostasis by bridging misfolded and aggregated proteins to the HSP70 machinery and to the degradative pathways, facilitating refolding or clearance of the potentially toxic proteins. Because of its activity against proteostasis alteration, the chaperone system plays a relevant role in the protection against proteotoxicity in MNDs. Here, we discuss the role of HSPBs in MNDs and which HSPBs may represent a valid target for therapeutic purposes.
Collapse
|
9
|
Heterogeneous Clinical Phenotypes of dHMN Caused by Mutation in HSPB1 Gene: A Case Series. Biomolecules 2022; 12:biom12101382. [PMID: 36291591 PMCID: PMC9599773 DOI: 10.3390/biom12101382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/07/2022] [Accepted: 09/22/2022] [Indexed: 11/21/2022] Open
Abstract
Mutations in HSPB1 are known to cause Charcot-Marie-Tooth disease type 2F (CMT2F) and distal hereditary motor neuropathy (dHMN). In this study, we presented three patients with mutation in HSPB1 who were diagnosed with dHMN. Proband 1 was a 14-year-old male with progressive bilateral lower limb weakness and walking difficulty for four years. Proband 2 was a 65-year-old male with chronic lower limb weakness and restless legs syndrome from the age of 51. Proband 3 was a 50-year-old female with progressive weakness, lower limbs atrophy from the age of 44. The nerve conduction studies (NCS) suggested axonal degeneration of the peripheral motor nerves and needle electromyography (EMG) revealed chronic neurogenic changes in probands. Open sural nerve biopsy for proband 2 and the mother of proband 1 showed mild to moderate loss of myelinated nerve fibers with some nerve fiber regeneration. A novel p.V97L in HSPB1 was identified in proband 3, the other two variants (p.P182A and p.R127W) in HSPB1 have been reported previously. The functional studies showed that expressing mutant p.V97L HSPB1 in SH-SY5Y cells displayed a decreased cell activity and increased apoptosis under stress condition. Our study expands the clinical phenotypic spectrum and etiological spectrum of HSPB1 mutation.
Collapse
|
10
|
Aghazadeh N, Beilankouhi EAV, Fakhri F, Gargari MK, Bahari P, Moghadami A, Khodabandeh Z, Valilo M. Involvement of heat shock proteins and parkin/α-synuclein axis in Parkinson's disease. Mol Biol Rep 2022; 49:11061-11070. [PMID: 36097120 DOI: 10.1007/s11033-022-07900-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurological diseases, next only to Alzheimer's disease (AD) in terms of prevalence. It afflicts about 2-3% of individuals over 65 years old. The etiology of PD is unknown and several environmental and genetic factors are involved. From a pathological point of view, PD is characterized by the loss of dopaminergic neurons in the substantia nigra, which causes the abnormal accumulation of α-synuclein (α-syn) (a component of Lewy bodies), which subsequently interact with heat shock proteins (HSPs), leading to apoptosis. Apoptosis is a vital pathway for establishing homeostasis in body tissues, which is regulated by pro-apoptotic and anti-apoptotic factors. Recent findings have shown that HSPs, especially HSP27 and HSP70, play a pivotal role in regulating apoptosis by influencing the factors involved in the apoptosis pathway. Moreover, it has been reported that the expression of these HSPs in the nervous system is high. Apart from this finding, investigations have suggested that HSP27 and HSP70 (related to parkin) show a potent protective and anti-apoptotic impact against the damaging outcomes of mutant α-syn toxicity to nerve cells. Therefore, in this study, we aimed to investigate the relationship between these HSPs and apoptosis in patients with PD.
Collapse
Affiliation(s)
- Nina Aghazadeh
- Department of biology, Islamic Azad University, Tabriz, Iran
| | | | - Farima Fakhri
- Research Institute for Neuroscience, Kerman University of Medical Sciences, Kerman, Iran
| | - Morad Kohandel Gargari
- Faculty of Medicine, Imamreza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Bahari
- Department of Clinical Biochemistry, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Aliasghar Moghadami
- Department of Clinical Biochemistry and Medical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zhila Khodabandeh
- Department of Biology, Faculty of Science, Urmia University, Urmia, Iran
| | - Mohammad Valilo
- Department of Clinical Biochemistry and Medical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Bluhm A, Schrempel S, Schilling S, von Hörsten S, Schulze A, Roßner S, Hartlage-Rübsamen M. Immunohistochemical Demonstration of the pGlu79 α-Synuclein Fragment in Alzheimer’s Disease and Its Tg2576 Mouse Model. Biomolecules 2022; 12:biom12071006. [PMID: 35883562 PMCID: PMC9312983 DOI: 10.3390/biom12071006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/14/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023] Open
Abstract
The deposition of β-amyloid peptides and of α-synuclein proteins is a neuropathological hallmark in the brains of Alzheimer’s disease (AD) and Parkinson’s disease (PD) subjects, respectively. However, there is accumulative evidence that both proteins are not exclusive for their clinical entity but instead co-exist and interact with each other. Here, we investigated the presence of a newly identified, pyroglutamate79-modified α-synuclein variant (pGlu79-aSyn)—along with the enzyme matrix metalloproteinase-3 (MMP-3) and glutaminyl cyclase (QC) implicated in its formation—in AD and in the transgenic Tg2576 AD mouse model. In the human brain, pGlu79-aSyn was detected in cortical pyramidal neurons, with more distinct labeling in AD compared to control brain tissue. Using immunohistochemical double and triple labelings and confocal laser scanning microscopy, we demonstrate an association of pGlu79-aSyn, MMP-3 and QC with β-amyloid plaques. In addition, pGlu79-aSyn and QC were present in amyloid plaque-associated reactive astrocytes that were also immunoreactive for the chaperone heat shock protein 27 (HSP27). Our data are consistent for the transgenic mouse model and the human clinical condition. We conclude that pGlu79-aSyn can be generated extracellularly or within reactive astrocytes, accumulates in proximity to β-amyloid plaques and induces an astrocytic protein unfolding mechanism involving HSP27.
Collapse
Affiliation(s)
- Alexandra Bluhm
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (Sa.S.); (M.H.-R.)
| | - Sarah Schrempel
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (Sa.S.); (M.H.-R.)
| | - Stephan Schilling
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, 06120 Halle (Saale), Germany; (S.S.); (A.S.)
- Faculty of Applied Biosciences and Process Engineering, Anhalt University of Applied Sciences, 06366 Köthen, Germany
| | - Stephan von Hörsten
- Department for Experimental Therapy, University Clinics Erlangen and Preclinical Experimental Center, University of Erlangen-Nuremberg, 91054 Erlangen, Germany;
| | - Anja Schulze
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, 06120 Halle (Saale), Germany; (S.S.); (A.S.)
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (Sa.S.); (M.H.-R.)
- Correspondence: ; Tel.: +49-341-9725758
| | - Maike Hartlage-Rübsamen
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; (A.B.); (Sa.S.); (M.H.-R.)
| |
Collapse
|
12
|
Tedesco B, Cristofani R, Ferrari V, Cozzi M, Rusmini P, Casarotto E, Chierichetti M, Mina F, Galbiati M, Piccolella M, Crippa V, Poletti A. Insights on Human Small Heat Shock Proteins and Their Alterations in Diseases. Front Mol Biosci 2022; 9:842149. [PMID: 35281256 PMCID: PMC8913478 DOI: 10.3389/fmolb.2022.842149] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
The family of the human small Heat Shock Proteins (HSPBs) consists of ten members of chaperones (HSPB1-HSPB10), characterized by a low molecular weight and capable of dimerization and oligomerization forming large homo- or hetero-complexes. All HSPBs possess a highly conserved centrally located α-crystallin domain and poorly conserved N- and C-terminal domains. The main feature of HSPBs is to exert cytoprotective functions by preserving proteostasis, assuring the structural maintenance of the cytoskeleton and acting in response to cellular stresses and apoptosis. HSPBs take part in cell homeostasis by acting as holdases, which is the ability to interact with a substrate preventing its aggregation. In addition, HSPBs cooperate in substrates refolding driven by other chaperones or, alternatively, promote substrate routing to degradation. Notably, while some HSPBs are ubiquitously expressed, others show peculiar tissue-specific expression. Cardiac muscle, skeletal muscle and neurons show high expression levels for a wide variety of HSPBs. Indeed, most of the mutations identified in HSPBs are associated to cardiomyopathies, myopathies, and motor neuropathies. Instead, mutations in HSPB4 and HSPB5, which are also expressed in lens, have been associated with cataract. Mutations of HSPBs family members encompass base substitutions, insertions, and deletions, resulting in single amino acid substitutions or in the generation of truncated or elongated proteins. This review will provide an updated overview of disease-related mutations in HSPBs focusing on the structural and biochemical effects of mutations and their functional consequences.
Collapse
Affiliation(s)
- B. Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - R. Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - V. Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - P. Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - E. Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - F. Mina
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - V. Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - A. Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
- *Correspondence: A. Poletti,
| |
Collapse
|
13
|
Dynamics of heat shock proteins and heat shock factor expression during heat stress in daughter workers in pre-heat-treated (rapid heat hardening) Apis mellifera mother queens. J Therm Biol 2022; 104:103194. [DOI: 10.1016/j.jtherbio.2022.103194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 01/08/2022] [Accepted: 01/21/2022] [Indexed: 11/24/2022]
|
14
|
Molecular chaperones and Parkinson's disease. Neurobiol Dis 2021; 160:105527. [PMID: 34626793 DOI: 10.1016/j.nbd.2021.105527] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 12/27/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive death of dopaminergic neurons in the substantia nigra and the formation of Lewy bodies (LBs). Mutations in PD-related genes lead to neuronal pathogenesis through various mechanisms, with known examples including SNCA/α-synuclein (PAKR1), Parkin (PARK2), PINK1 (PARK6), DJ-1 (PARK7), and LRRK2 (PARK8). Molecular chaperones/co-chaperones are proteins that aid the folding of other proteins into a functionally active conformation. It has been demonstrated that chaperones/co-chaperones interact with PD-related proteins and regulate their function in PD. HSP70, HSP90 and small heat shock proteins can prevent neurodegeneration by regulating α-syn misfolding, oligomerization and aggregation. The function of chaperones is regulated by co-chaperones such as HSP110, HSP40, HOP, CHIP, and BAG family proteins. Parkin, PINK1 and DJ-1 are PD-related proteins which are associated with mitochondrial function. Molecular chaperones regulate mitochondrial function and protein homeostasis by interacting with these PD-related proteins. This review discusses critical molecular chaperones/co-chaperones and PD-related proteins which contribute to the pathogenesis of PD, hoping to provide new molecular targets for therapeutic interventions to thwart the disease progression instead of only bringing symptomatic relief. Moreover, appreciating the critical role of chaperones in PD can also help us screen efficient biomarkers to identify PD at an early stage.
Collapse
|
15
|
Vahidinia Z, Mahdavi E, Talaei SA, Naderian H, Tamtaji A, Haddad Kashani H, Beyer C, Azami Tameh A. The effect of female sex hormones on Hsp27 phosphorylation and histological changes in prefrontal cortex after tMCAO. Pathol Res Pract 2021; 221:153415. [PMID: 33857717 DOI: 10.1016/j.prp.2021.153415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/13/2021] [Accepted: 03/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Female sex hormones are protective factors against many neurological disorders such as brain ischemia. Heat shock protein like HSP27 is activated after tissue injury. The main purpose of the present study is to determine the effect of a combined estrogen / progesterone cocktail on the morphology of astrocytes, neurons and Hsp27 phosphorylation after cerebral ischemia. METHODS One hour after the MCAO induction, a single dose of estrogen and progesterone was injected. The infarct volume was calculated by TTC staining 24 h after ischemia. Immunohistochemistry was used to show the effects of estrogen and progesterone on astrocyte and neuron morphology, as well as the Western blot technique used for the quantitation of phosphorylated Hsp27. RESULTS The combined dose of estrogen and progesterone significantly decreased astrocytosis after ischemia and increased neuron survival. There was a large increase in Hsp27 phosphorylation in the penumbra ischemic region after stroke, which was significantly reduced by hormone therapy. CONCLUSION Our results indicate that the neuroprotective effect of neurosteroids in the brain may be due to the modulation of heat shock proteins.
Collapse
Affiliation(s)
- Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Elham Mahdavi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Homayoun Naderian
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Aboutaleb Tamtaji
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Haddad Kashani
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
16
|
Chaudhury S, Keegan BM, Blagg BSJ. The role and therapeutic potential of Hsp90, Hsp70, and smaller heat shock proteins in peripheral and central neuropathies. Med Res Rev 2021; 41:202-222. [PMID: 32844464 PMCID: PMC8485878 DOI: 10.1002/med.21729] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022]
Abstract
Heat shock proteins (Hsps) are molecular chaperones that also play important roles in the activation of the heat shock response (HSR). The HSR is an evolutionary conserved and protective mechanism that is used to counter abnormal physiological conditions, stressors, and disease states, such as those exemplified in cancer and/or neurodegeneration. In normal cells, heat shock factor-1 (HSF-1), the transcription factor that regulates the HSR, remains in a dormant multiprotein complex that is formed upon association with chaperones (Hsp90, Hsp70, etc.), co-chaperones, and client proteins. However, under cellular stress, HSF-1 dissociates from Hsp90 and induces the transcriptional upregulation of Hsp70 to afford protection against the encountered cellular stress. As a consequence of both peripheral and central neuropathies, cellular stress occurs and results in the accumulation of unfolded and/or misfolded proteins, which can be counterbalanced by activation of the HSR. Since Hsp90 is the primary regulator of the HSR, modulation of Hsp90 by small molecules represents an attractive therapeutic approach against both peripheral and central neuropathies.
Collapse
Affiliation(s)
- Subhabrata Chaudhury
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana, USA
| | - Bradley M Keegan
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana, USA
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
17
|
Saikumar J, Byrns CN, Hemphill M, Meaney DF, Bonini NM. Dynamic neural and glial responses of a head-specific model for traumatic brain injury in Drosophila. Proc Natl Acad Sci U S A 2020; 117:17269-17277. [PMID: 32611818 PMCID: PMC7382229 DOI: 10.1073/pnas.2003909117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is the strongest environmental risk factor for the accelerated development of neurodegenerative diseases. There are currently no therapeutics to address this due to lack of insight into mechanisms of injury progression, which are challenging to study in mammalian models. Here, we have developed and extensively characterized a head-specific approach to TBI in Drosophila, a powerful genetic system that shares many conserved genes and pathways with humans. The Drosophila TBI (dTBI) device inflicts mild, moderate, or severe brain trauma by precise compression of the head using a piezoelectric actuator. Head-injured animals display features characteristic of mammalian TBI, including severity-dependent ataxia, life span reduction, and brain degeneration. Severe dTBI is associated with cognitive decline and transient glial dysfunction, and stimulates antioxidant, proteasome, and chaperone activity. Moreover, genetic or environmental augmentation of the stress response protects from severe dTBI-induced brain degeneration and life span deficits. Together, these findings present a tunable, head-specific approach for TBI in Drosophila that recapitulates mammalian injury phenotypes and underscores the ability of the stress response to mitigate TBI-induced brain degeneration.
Collapse
Affiliation(s)
- Janani Saikumar
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - China N Byrns
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Matthew Hemphill
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104;
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
18
|
Vendredy L, Adriaenssens E, Timmerman V. Small heat shock proteins in neurodegenerative diseases. Cell Stress Chaperones 2020; 25:679-699. [PMID: 32323160 PMCID: PMC7332613 DOI: 10.1007/s12192-020-01101-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
Small heat shock proteins are ubiquitously expressed chaperones, yet mutations in some of them cause tissue-specific diseases. Here, we will discuss how small heat shock proteins give rise to neurodegenerative disorders themselves while we will also highlight how these proteins can fulfil protective functions in neurodegenerative disorders caused by protein aggregation. The first half of this paper will be focused on how mutations in HSPB1, HSPB3, and HSPB8 are linked to inherited peripheral neuropathies like Charcot-Marie-Tooth (CMT) disease and distal hereditary motor neuropathy (dHMN). The second part of the paper will discuss how small heat shock proteins are linked to neurodegenerative disorders like Alzheimer's, Parkinson's, and Huntington's disease.
Collapse
Affiliation(s)
- Leen Vendredy
- Department of Biomedical Sciences and Institute Born Bunge, Peripheral Neuropathy Research Group, University of Antwerp, Antwerp, Belgium
| | - Elias Adriaenssens
- Department of Biomedical Sciences and Institute Born Bunge, Peripheral Neuropathy Research Group, University of Antwerp, Antwerp, Belgium
| | - Vincent Timmerman
- Department of Biomedical Sciences and Institute Born Bunge, Peripheral Neuropathy Research Group, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
19
|
Vicente Miranda H, Chegão A, Oliveira MS, Fernandes Gomes B, Enguita FJ, Outeiro TF. Hsp27 reduces glycation-induced toxicity and aggregation of alpha-synuclein. FASEB J 2020; 34:6718-6728. [PMID: 32259355 DOI: 10.1096/fj.201902936r] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/02/2020] [Accepted: 03/12/2020] [Indexed: 01/20/2023]
Abstract
α-synuclein (aSyn) is a major player in Parkinson's disease and a group of other disorders collectively known as synucleinopathies, but the precise molecular mechanisms involved are still unclear. aSyn, as virtually all proteins, undergoes a series of posttranslational modifications during its lifetime, which can affect its biology and pathobiology. We recently showed that glycation of aSyn by methylglyoxal (MGO) potentiates its oligomerization and toxicity, induces dopaminergic neuronal cell loss in mice, and affects motor performance in flies. Small heat-shock proteins (sHsps) are molecular chaperones that facilitate the folding of proteins or target misfolded proteins for clearance. Importantly, sHsps were shown to prevent aSyn aggregation and cytotoxicity. Upon treating cells with increasing amounts of methylglyoxal, we found that the levels of Hsp27 decreased in a dose-dependent manner. Therefore, we hypothesized that restoring the levels of Hsp27 in glycating environments could alleviate the pathogenicity of aSyn. Consistently, we found that Hsp27 reduced MGO-induced aSyn aggregation in cells, leading to the formation of nontoxic aSyn species. Remarkably, increasing the levels of Hsp27 suppressed the deleterious effects induced by MGO. Our findings suggest that in glycating environments, the levels of Hsp27 are important for modulating the glycation-associated cellular pathologies in synucleinopathies.
Collapse
Affiliation(s)
- Hugo Vicente Miranda
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Ana Chegão
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Márcia S Oliveira
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Bárbara Fernandes Gomes
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Francisco J Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago Fleming Outeiro
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal.,Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
20
|
Dar KB, Bhat AH, Amin S, Reshi BA, Zargar MA, Masood A, Ganie SA. Elucidating Critical Proteinopathic Mechanisms and Potential Drug Targets in Neurodegeneration. Cell Mol Neurobiol 2020; 40:313-345. [PMID: 31584139 PMCID: PMC11449027 DOI: 10.1007/s10571-019-00741-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/06/2019] [Indexed: 12/18/2022]
Abstract
Neurodegeneration entails progressive loss of neuronal structure as well as function leading to cognitive failure, apathy, anxiety, irregular body movements, mood swing and ageing. Proteomic dysregulation is considered the key factor for neurodegeneration. Mechanisms involving deregulated processing of proteins such as amyloid beta (Aβ) oligomerization; tau hyperphosphorylation, prion misfolding; α-synuclein accumulation/lewy body formation, chaperone deregulation, acetylcholine depletion, adenosine 2A (A2A) receptor hyperactivation, secretase deregulation, leucine-rich repeat kinase 2 (LRRK2) mutation and mitochondrial proteinopathies have deeper implications in neurodegenerative disorders. Better understanding of such pathological mechanisms is pivotal for exploring crucial drug targets. Herein, we provide a comprehensive outlook about the diverse proteomic irregularities in Alzheimer's, Parkinson's and Creutzfeldt Jakob disease (CJD). We explicate the role of key neuroproteomic drug targets notably Aβ, tau, alpha synuclein, prions, secretases, acetylcholinesterase (AchE), LRRK2, molecular chaperones, A2A receptors, muscarinic acetylcholine receptors (mAchR), N-methyl-D-aspartate receptor (NMDAR), glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) and mitochondrial/oxidative stress-related proteins for combating neurodegeneration and associated cognitive and motor impairment. Cross talk between amyloidopathy, synucleinopathy, tauopathy and several other proteinopathies pinpoints the need to develop safe therapeutics with ability to strike multiple targets in the aetiology of the neurodegenerative disorders. Therapeutics like microtubule stabilisers, chaperones, kinase inhibitors, anti-aggregation agents and antibodies could serve promising regimens for treating neurodegeneration. However, drugs should be target specific, safe and able to penetrate blood-brain barrier.
Collapse
Affiliation(s)
- Khalid Bashir Dar
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Aashiq Hussain Bhat
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Shajrul Amin
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Bilal Ahmad Reshi
- Department of Biotechnology, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Mohammad Afzal Zargar
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Akbar Masood
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Showkat Ahmad Ganie
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India.
| |
Collapse
|
21
|
Verma S, Sharma S, Ranawat P, Nehru B. Modulatory Effects of Ginkgo biloba Against Amyloid Aggregation Through Induction of Heat Shock Proteins in Aluminium Induced Neurotoxicity. Neurochem Res 2020; 45:465-490. [PMID: 31894463 DOI: 10.1007/s11064-019-02940-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 11/27/2019] [Accepted: 12/18/2019] [Indexed: 01/04/2023]
Abstract
Protein misfolding and aggregation of amyloid beta (Aβ) peptide, as well as formation of neurofibrillary tangles (NFTs) are the signature hallmarks of Alzheimer's disease (AD) pathology. To prevent this, molecular chaperones come into play as they facilitate the refolding of the misfolded proteins and cell protection under stress. Here, we have evaluated the possible effects of Ginkgo biloba (GBE) against aggregation of the Aβ through activation of heat shock proteins (HSPs) in the Aluminium (Al) induced AD based model. GBE (100 mg/kg body weight) was administered per oral to the female SD rats in conjunction with intraperitoneal (i.p.) injection of Al lactate (10 mg/kg body weight) for six weeks. Pretreated animals were administered GBE for additional two weeks prior to any exposure of Al. GBE administration resulted in decrease in Aβ aggregation, ubiquitin deposition, accompanying a significant decline in APP & Tau protein hyperphosphorylation which can be attributed to activation of Heat shock factor (HSF-1) and upregulation in the protein expression of HSPs. Histopathological investigation studies have also shown the decrease in aggregation of Aβ peptide by GBE administration. Additionally, the decrease in ROS levels and Aβ aggregation by GBE administration prohibited the decline in the neurotransmitter levels and monoamine oxidase levels in hippocampus and cortex. This further caused improvement in learning and memory of the animals. In conclusion, our results indicate that GBE prevents the symptoms of Al induced AD like pathophysiology by upregulating the HSPs levels and decreasing the aggregation load.
Collapse
Affiliation(s)
- Sonia Verma
- Department of Biophysics, South Campus, Panjab University, Chandigarh, 160014, India
| | - Sheetal Sharma
- Department of Biophysics, South Campus, Panjab University, Chandigarh, 160014, India
| | - Pavitra Ranawat
- Department of Biophysics, South Campus, Panjab University, Chandigarh, 160014, India
| | - Bimla Nehru
- Department of Biophysics, South Campus, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
22
|
Lázaro DF, Outeiro TF. The Interplay Between Proteostasis Systems and Parkinson’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:223-236. [DOI: 10.1007/978-3-030-38266-7_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
23
|
Heat shock protein signaling in brain ischemia and injury. Neurosci Lett 2019; 715:134642. [PMID: 31759081 DOI: 10.1016/j.neulet.2019.134642] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 12/28/2022]
Abstract
Heat shock proteins (HSPs) are chaperones that catalyze the refolding of denatured proteins. In addition to their ability to prevent protein denaturation and aggregation, the HSPs have also been shown to modulate many signaling pathways. Among HSPs, the inducible 70 kDa HSP (HSP70) has especially been shown to improve neurological outcome in experimental models of brain ischemia and injury. HSP70 can modulate various aspects of the programmed cell death pathways and inflammation. This review will focus on potential mechanisms of the neuroprotective effects of HSP70 in stroke and brain trauma models. We also comment on potential ways in which HSP70 could be translated into clinical therapies.
Collapse
|
24
|
Jia C, Ma X, Liu Z, Gu J, Zhang X, Li D, Zhang S. Different Heat Shock Proteins Bind α-Synuclein With Distinct Mechanisms and Synergistically Prevent Its Amyloid Aggregation. Front Neurosci 2019; 13:1124. [PMID: 31749672 PMCID: PMC6842937 DOI: 10.3389/fnins.2019.01124] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 10/04/2019] [Indexed: 01/08/2023] Open
Abstract
α-Synuclein (α-Syn) forms pathological amyloid aggregates deposited in Lewy bodies and Lewy neurites in the brain of Parkinson's disease (PD) patients. Heat shock proteins (Hsps) are the major components of the cellular chaperone network, which are responsible for preventing proteins from amyloid aggregation. Different Hsps were reported to interact with α-syn. However, the underlying mechanism of the interplay between α-syn and different Hsps remains unclear. Here, by combing NMR spectroscopy, electron microscope and other biochemical approaches, we systemically investigated the interaction between α-syn and three Hsps from different families including Hsp27, HDJ1, and Hsp104. We found that all three Hsps can weakly bind to α-syn and inhibit it from amyloid aggregation. Intriguingly, different Hsps recognize distinct regions of α-syn monomer, and act synergistically in chaperoning α-syn from fibril formation in sub-stoichiometry. Our results revealed the diverse binding mechanisms employed by different Hsps to tackle α-syn, and suggested that different Hsps form a network for cooperatively chaperoning α-syn from pathological aggregation.
Collapse
Affiliation(s)
- Chunyu Jia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.,Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, University of the Chinese Academy of Sciences, Beijing, China
| | - Xiaojuan Ma
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.,Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, University of the Chinese Academy of Sciences, Beijing, China
| | - Zhenying Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.,Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, University of the Chinese Academy of Sciences, Beijing, China
| | - Jinge Gu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.,Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, University of the Chinese Academy of Sciences, Beijing, China
| | - Xiang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.,Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, University of the Chinese Academy of Sciences, Beijing, China
| | - Dan Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Shengnan Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
25
|
Dukay B, Csoboz B, Tóth ME. Heat-Shock Proteins in Neuroinflammation. Front Pharmacol 2019; 10:920. [PMID: 31507418 PMCID: PMC6718606 DOI: 10.3389/fphar.2019.00920] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/22/2019] [Indexed: 01/01/2023] Open
Abstract
The heat-shock response, one of the main pro-survival mechanisms of a living organism, has evolved as the biochemical response of cells to cope with heat stress. The most well-characterized aspect of the heat-shock response is the accumulation of a conserved set of proteins termed heat-shock proteins (HSPs). HSPs are key players in protein homeostasis acting as chaperones by aiding the folding and assembly of nascent proteins and protecting against protein aggregation. HSPs have been associated with neurological diseases in the context of their chaperone activity, as they were found to suppress the aggregation of misfolded toxic proteins. In recent times, HSPs have proven to have functions apart from the classical molecular chaperoning in that they play a role in a wider scale of neurological disorders by modulating neuronal survival, inflammation, and disease-specific signaling processes. HSPs are gaining importance based on their ability to fine-tune inflammation and act as immune modulators in various bodily fluids. However, their effect on neuroinflammation processes is not yet fully understood. In this review, we summarize the role of neuroinflammation in acute and chronic pathological conditions affecting the brain. Moreover, we seek to explore the existing literature on HSP-mediated inflammatory function within the central nervous system and compare the function of these proteins when they are localized intracellularly compared to being present in the extracellular milieu.
Collapse
Affiliation(s)
- Brigitta Dukay
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Bálint Csoboz
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| |
Collapse
|
26
|
Charcot-Marie-Tooth 2F (Hsp27 mutations): A review. Neurobiol Dis 2019; 130:104505. [PMID: 31212070 DOI: 10.1016/j.nbd.2019.104505] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
Charcot-Marie-Tooth disease is a commonly inherited form of neuropathy. Although named over 100 years ago, identification of subtypes of Charcot-Marie-Tooth has rapidly expanded in the preceding decades with the advancement of genetic sequencing, including type 2F (CMT2F), due to mutations in heat shock protein 27 (Hsp27). However, despite CMT being one of the most common inherited neurological diseases, definitive mechanistic models of pathology and effective treatments for CMT2F are lacking. This review extensively profiles the published literature on CMT2F and distal hereditary motor neuropathy II (dHMN II), a similar neuropathy with exclusively motor symptoms that is also due to mutations in Hsp27. This includes a review of case reports and sequencing studies detailing disease course. Included are tables listing of all known published mutations of Hsp27 that cause symptoms of CMT2F and dHMN II. Furthermore, pathological mechanisms are assessed. While many groups have established pathologies relating to defective chaperone function, cellular neurofilament and microtubule structure and function, and mitochondrial and metabolic dysfunction, there are still discrepancies in results between different model systems. Moreover, initial mouse models have also produced promising results with similar phenotypes to humans, however discrepancies still exist. Both patient-focused and scientific studies have demonstrated variability in phenotypes even considering specific mutations. Given the clinical heterogeneity in presentation, CMT2F and dHMN II likely result from similar pathological mechanisms of the same general disease process that may present distinctly due to other genetic and environment influences. Determining how these influences exert their effects to produce pathology contributing to the disease phenotype will be a major future challenge ahead in the field.
Collapse
|
27
|
Exosomal secretion of α-synuclein as protective mechanism after upstream blockage of macroautophagy. Cell Death Dis 2018; 9:757. [PMID: 29988147 PMCID: PMC6037700 DOI: 10.1038/s41419-018-0816-2] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 01/16/2023]
Abstract
Accumulation of pathological α-synuclein aggregates plays a major role in Parkinson’s disease. Macroautophagy is a mechanism to degrade intracellular protein aggregates by wrapping them into autophagosomes, followed by fusion with lysosomes. We had previously shown that pharmacological activation of macroautophagy protects against α-synuclein-induced toxicity in human neurons. Here, we hypothesized that inhibition of macroautophagy would aggravate α-synuclein-induced cell death. Unexpectedly, inhibition of autophagosome formation by silencing of ATG5 protected from α-synuclein-induced toxicity. Therefore, we studied alternative cellular mechanisms to compensate for the loss of macroautophagy. ATG5 silencing did not affect the ubiquitin–proteasome system, chaperone systems, chaperone-mediated autophagy, or the unfolded protein response. However, ATG5 silencing increased the secretion of α-synuclein via exosomes. Blocking exosomal secretion exacerbated α-synuclein-induced cell death. We conclude that exosomal secretion of α-synuclein is increased after impaired formation of autophagosomes to reduce the intracellular α-synuclein burden. This compensatory mechanism prevents α-synuclein-induced neuronal cell death.
Collapse
|
28
|
Kim JH, Jung YS, Kim JW, Ha MS, Ha SM, Kim DY. Effects of aquatic and land-based exercises on amyloid beta, heat shock protein 27, and pulse wave velocity in elderly women. Exp Gerontol 2018; 108:62-68. [PMID: 29604402 DOI: 10.1016/j.exger.2018.03.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/12/2018] [Accepted: 03/27/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Alzheimer's disease is a neurodegenerative brain disease resulting from the deterioration of neuronal cells and vascular dementia, the latter of which results from cerebrovascular disorders. Exercise is effective in preventing and treating degenerative brain diseases as it activates blood flow to the brain, increases nerve production in the hippocampus, and promotes the expression of synaptic plasticity-related proteins. Therefore, this study investigated the effects of 16-week aquatic and land-based exercise programs on amyloid beta (Aβ), heat shock protein (HSP) 27 levels, and pulse wave velocity (PWV). MATERIALS AND METHODS Forty elderly women, aged 60-70 years, voluntarily participated in the study. They were divided into control (n = 12), aquatic exercise (n = 14), and land-based exercise groups (n = 14). The variables of amyloid beta, heat shock protein 27, and pulse wave velocity were measured in all the participants before and after the 16-week study. RESULTS Significantly higher levels of serum HSP27 (p < 0.05) and significantly lower levels of vascular elasticity (p < 0.05) were found in the aquatic exercise group after 16 weeks of exercise compared with the control group. Aβ did not significantly differ between groups. Thirty minutes after the first exercise, Aβ in the aquatic exercise group (p < 0.01) and HSP27 in the land-based exercise group (p < 0.05) were significantly higher than the corresponding levels in the resting condition before exercise. 30 min after the last exercise, Aβ (p < 0.01) and HSP27 (p < 0.05) were significantly higher. CONCLUSIONS Aquatic and land-based exercises increased serum Aβ and HSP27 and decreased pulse wave velocity. Thus, they may play a positive role in the prevention of degenerative brain diseases and improvement of brain function in elderly people.
Collapse
Affiliation(s)
- Ji-Hyeon Kim
- Department of Physical Education, Pusan National University, Busan, South Korea
| | - Young-Suk Jung
- Department of Pharmacy, Pusan National University, Busan, South Korea
| | - Jong-Won Kim
- Department of Physical Education, Busan National University of Education, Busan, South Korea
| | - Min-Seong Ha
- Department of Physical Education, Pusan National University, Busan, South Korea
| | - Soo-Min Ha
- Department of Physical Education, Pusan National University, Busan, South Korea
| | - Do-Yeon Kim
- Department of Physical Education, Pusan National University, Busan, South Korea.
| |
Collapse
|
29
|
Penke B, Bogár F, Crul T, Sántha M, Tóth ME, Vígh L. Heat Shock Proteins and Autophagy Pathways in Neuroprotection: from Molecular Bases to Pharmacological Interventions. Int J Mol Sci 2018; 19:E325. [PMID: 29361800 PMCID: PMC5796267 DOI: 10.3390/ijms19010325] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases (NDDs) such as Alzheimer's disease, Parkinson's disease and Huntington's disease (HD), amyotrophic lateral sclerosis, and prion diseases are all characterized by the accumulation of protein aggregates (amyloids) into inclusions and/or plaques. The ubiquitous presence of amyloids in NDDs suggests the involvement of disturbed protein homeostasis (proteostasis) in the underlying pathomechanisms. This review summarizes specific mechanisms that maintain proteostasis, including molecular chaperons, the ubiquitin-proteasome system (UPS), endoplasmic reticulum associated degradation (ERAD), and different autophagic pathways (chaperon mediated-, micro-, and macro-autophagy). The role of heat shock proteins (Hsps) in cellular quality control and degradation of pathogenic proteins is reviewed. Finally, putative therapeutic strategies for efficient removal of cytotoxic proteins from neurons and design of new therapeutic targets against the progression of NDDs are discussed.
Collapse
Affiliation(s)
- Botond Penke
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Dóm Square 8, Hungary.
| | - Ferenc Bogár
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Dóm Square 8, Hungary.
- MTA-SZTE Biomimetic Systems Research Group, University of Szeged, H-6720 Szeged, Dóm Square 8, Hungary.
| | - Tim Crul
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62, Hungary.
| | - Miklós Sántha
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62, Hungary.
| | - Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62, Hungary.
| | - László Vígh
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, H-6726 Szeged, Temesvári krt. 62, Hungary.
| |
Collapse
|
30
|
Maiti P, Manna J, Dunbar GL. Current understanding of the molecular mechanisms in Parkinson's disease: Targets for potential treatments. Transl Neurodegener 2017; 6:28. [PMID: 29090092 PMCID: PMC5655877 DOI: 10.1186/s40035-017-0099-z] [Citation(s) in RCA: 328] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022] Open
Abstract
Gradual degeneration and loss of dopaminergic neurons in the substantia nigra, pars compacta and subsequent reduction of dopamine levels in striatum are associated with motor deficits that characterize Parkinson’s disease (PD). In addition, half of the PD patients also exhibit frontostriatal-mediated executive dysfunction, including deficits in attention, short-term working memory, speed of mental processing, and impulsivity. The most commonly used treatments for PD are only partially or transiently effective and are available or applicable to a minority of patients. Because, these therapies neither restore the lost or degenerated dopaminergic neurons, nor prevent or delay the disease progression, the need for more effective therapeutics is critical. In this review, we provide a comprehensive overview of the current understanding of the molecular signaling pathways involved in PD, particularly within the context of how genetic and environmental factors contribute to the initiation and progression of this disease. The involvement of molecular chaperones, autophagy-lysosomal pathways, and proteasome systems in PD are also highlighted. In addition, emerging therapies, including pharmacological manipulations, surgical procedures, stem cell transplantation, gene therapy, as well as complementary, supportive and rehabilitation therapies to prevent or delay the progression of this complex disease are reviewed.
Collapse
Affiliation(s)
- Panchanan Maiti
- Field Neurosciences Institute Laboratory for Restorative Neurology, Mt. Pleasant, MI 48859 USA.,Program in Neuroscience, Mt. Pleasant, MI 48859 USA.,Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859 USA.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI 48604 USA.,Department of Biology, Saginaw Valley State University, Saginaw, MI 48604 USA
| | - Jayeeta Manna
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38105 USA
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Mt. Pleasant, MI 48859 USA.,Program in Neuroscience, Mt. Pleasant, MI 48859 USA.,Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859 USA.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI 48604 USA
| |
Collapse
|
31
|
Cox D, Ecroyd H. The small heat shock proteins αB-crystallin (HSPB5) and Hsp27 (HSPB1) inhibit the intracellular aggregation of α-synuclein. Cell Stress Chaperones 2017; 22:589-600. [PMID: 28337642 PMCID: PMC5465035 DOI: 10.1007/s12192-017-0785-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 01/11/2023] Open
Abstract
Protein homeostasis, or proteostasis, is the process of maintaining the conformational and functional integrity of the proteome. Proteostasis is preserved in the face of stress by a complex network of cellular machinery, including the small heat shock molecular chaperone proteins (sHsps), which act to inhibit the aggregation and deposition of misfolded protein intermediates. Despite this, the pathogenesis of several neurodegenerative diseases has been inextricably linked with the amyloid fibrillar aggregation and deposition of α-synuclein (α-syn). The sHsps are potent inhibitors of α-syn aggregation in vitro. However, the limited availability of a robust, cell-based model of α-syn aggregation has, thus far, restricted evaluation of sHsp efficacy in the cellular context. As such, this work sought to establish a robust model of intracellular α-syn aggregation using Neuro-2a cells. Aggregation of α-syn was found to be sensitive to inhibition of autophagy and the proteasome, resulting in a significant increase in the proportion of cells containing α-syn inclusions. This model was then used to evaluate the capacity of the sHsps, αB-c and Hsp27, to prevent α-syn aggregation in cells. To do so, we used bicistronic expression plasmids to express the sHsps. Unlike traditional fluorescent fusion constructs, these bicistronic expression plasmids enable only individual transfected cells expressing the sHsps (via expression of the fluorescent reporter) to be analysed, but without the need to tag the sHsp, which can affect its oligomeric structure and chaperone activity. Overexpression of both αB-c and Hsp27 significantly reduced the intracellular aggregation of α-syn. Thus, these findings suggest that overexpressing or boosting the activity of sHsps may be a way of preventing amyloid fibrillar aggregation of α-syn in the context of neurodegenerative disease.
Collapse
Affiliation(s)
- Dezerae Cox
- Illawarra Health and Medical Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia
- School of Biological Sciences, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Heath Ecroyd
- Illawarra Health and Medical Research Institute, University of Wollongong, Northfields Avenue, Wollongong, NSW, 2522, Australia.
- School of Biological Sciences, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
32
|
Banerjee PR, Moosa MM, Deniz AA. Two-Dimensional Crowding Uncovers a Hidden Conformation of α-Synuclein. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201606963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Priya R. Banerjee
- Department of Integrative Structural and Computational Biology; The Scripps Research Institute; La Jolla CA 92037 USA
| | - Mahdi Muhammad Moosa
- Department of Integrative Structural and Computational Biology; The Scripps Research Institute; La Jolla CA 92037 USA
| | - Ashok A. Deniz
- Department of Integrative Structural and Computational Biology; The Scripps Research Institute; La Jolla CA 92037 USA
| |
Collapse
|
33
|
Nafar F, Williams JB, Mearow KM. Astrocytes release HspB1 in response to amyloid-β exposure in vitro. J Alzheimers Dis 2016; 49:251-63. [PMID: 26444769 DOI: 10.3233/jad-150317] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although heat shock proteins are thought to function primarily as intracellular chaperones, the release and potential extracellular functions of heat shock proteins have been the focus of an increasing number of studies. Our particular interest is HspB1 (Hsp25/27) and as astrocytes are an in vivo source of HspB1 it is a reasonable possibility they could release HspB1 in response to local stresses. Using primary cultures of rat cortical astrocytes, we investigated the extracellular release of HspB1 with exposure to amyloid-β (Aβ). In order to assess potential mechanisms of release, we cotreated the cells with compounds that can modulate protein secretion including Brefeldin A, Methyl β-cyclodextrin, and MAP kinase inhibitors. Exposure to Aβ (0.1, 1.0, 2.0 μM) for 24-48 h resulted in a selective release of HspB1 that was insensitive to BFA treatment; none of the other inhibitors had any detectable influence. Protease protection assays indicated that some of the released HspB1 was associated with a membrane bound fraction, and analysis of exosomal preparations indicated the presence of HspB1 in exosomes. Finally, immunoprecipitation experiments demonstrated that the extracellular HspB1 was able to interact with extracellular Aβ. In summary, Aβ can stimulate release of HspB1 from astrocytes, this release is insensitive to Golgi or lipid raft disruption, and HspB1 can be found either free in the medium or associated with exosomes. This release suggests that there is a potential for extracellular HspB1 to be able to bind and sequester extracellular Aβ.
Collapse
|
34
|
Banerjee PR, Moosa MM, Deniz AA. Two-Dimensional Crowding Uncovers a Hidden Conformation of α-Synuclein. Angew Chem Int Ed Engl 2016; 55:12789-12792. [PMID: 27612332 DOI: 10.1002/anie.201606963] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Indexed: 11/09/2022]
Abstract
The intrinsically disordered protein (IDP), α-synuclein (αS), is well-known for phospholipid membrane binding-coupled folding into tunable helical conformers. Here, using single-molecule experiments in conjunction with ensemble assays and a theoretical model, we present a unique case demonstrating that the interaction-folding landscape of αS can be tuned by two-dimensional (2D) crowding through simultaneous binding of a second protein on the bilayer surface. Unexpectedly, the experimental data show a clear deviation from a simple competitive inhibition model, but are consistent with a bimodal inhibition mechanism wherein membrane binding of a second protein (a membrane interacting chaperone, Hsp27, in this case) differentially inhibits two distinct modules of αS-membrane interaction. As a consequence, αS molecules are forced to access a hidden conformational state on the phospholipid bilayer in which only the higher-affinity module remains membrane-bound. Our results demonstrate that macromolecular crowding in two dimensions can play a significant role in shaping the conformational landscape of membrane-binding IDPs with multiple binding modes.
Collapse
Affiliation(s)
- Priya R Banerjee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Mahdi Muhammad Moosa
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Ashok A Deniz
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
35
|
Chang KH, Lin CH, Chen HC, Huang HY, Chen SL, Lin TH, Ramesh C, Huang CC, Fung HC, Wu YR, Huang HJ, Lee-Chen GJ, Hsieh-Li HM, Yao CF. The Potential of Indole/Indolylquinoline Compounds in Tau Misfolding Reduction by Enhancement of HSPB1. CNS Neurosci Ther 2016; 23:45-56. [PMID: 27424519 DOI: 10.1111/cns.12592] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 06/17/2016] [Accepted: 06/19/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Neurofibrillary tangles formed from tau misfolding have long been considered one of the pathological hallmarks of Alzheimer's disease (AD). The misfolding of tau in AD correlates with the clinical progression of AD and inhibition or reversal of tau misfolding may protect the affected neurons. METHODS We generated 293 and SH-SY5Y cells expressing DsRed-tagged pro-aggregation mutant of repeat domain of tau (ΔK280 tauRD ) to test indole/indolylquinoline derivatives for reducing tau misfolding and neuroprotection. RESULTS Four of the 10 derivatives tested displayed good misfolding-inhibitory effects on Tet-On 293 cells. Among them, NC009-1 and NC009-7 enhanced heat-shock 27 kDa protein 1 (HSPB1) expression to increase ∆K280 tauRD -DsRed solubility and promoted neurite outgrowth in Tet-On SH-SY5Y cells. Knockdown of HSPB1 resulted in decreased ∆K280 tauRD -DsRed solubility and reduced neurite outgrowth, which were rescued by addition of NC009-1/NC009-7. Treatment with indole/indolylquinoline derivatives also improved neuronal cell viability and neurite outgrowth in mouse hippocampal primary culture under tau cytotoxicity. CONCLUSION Our results demonstrate how indole/indolylquinoline derivatives are likely to work in tau misfolding reduction, providing insight into the possible working mechanism of indole and indolylquinoline derivatives in AD treatment.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Hsin Lin
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hsuan-Chiang Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsin-Yu Huang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Shu-Ling Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Te-Hsien Lin
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chintakunta Ramesh
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Chin-Chang Huang
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hon-Chung Fung
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hei-Jen Huang
- Department of Nursing, Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ching-Fa Yao
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
36
|
Lin CH, Hsieh YS, Wu YR, Hsu CJ, Chen HC, Huang WH, Chang KH, Hsieh-Li HM, Su MT, Sun YC, Lee GC, Lee-Chen GJ. Identifying GSK-3β kinase inhibitors of Alzheimer's disease: Virtual screening, enzyme, and cell assays. Eur J Pharm Sci 2016; 89:11-9. [PMID: 27094783 DOI: 10.1016/j.ejps.2016.04.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 04/04/2016] [Accepted: 04/10/2016] [Indexed: 12/12/2022]
Abstract
Glycogen synthase kinase 3β (GSK-3β) is widely known as a critical target protein for treating Alzheimer's disease (AD). We utilized virtual screening to search databases for compounds with the potential to be used in drugs targeting GSK-3β kinase, and kinase as well as cell assays to investigate top-scored, selected compounds. Virtual screening of >1.1 million compounds in the ZINC and in-house databases was conducted using an optimized computational protocol in the docking program GOLD. Of the top-ranked compounds, 16 underwent a luminescent kinase assay and a cell assay using HEK293 cells expressing DsRed-tagged ΔK280 in the repeat domain of tau (tauRD). The compounds VB-003 (a potent GSK-3β inhibitor) and VB-008 (AM404, an anandamide transport inhibitor), with determined IC50 values of 0.25 and 5.4μM, respectively, were identified as reducing tau aggregation. Both compounds increased expression of phospho-GSK-3β (Ser9) and reduced endogenous tau phosphorylation at the sites of Ser202, Thr231, and Ser396. In the ∆K280 tauRD-DsRed SH-SY5Y cells, VB-008, but not VB-003, enhanced HSPB1 and GRP78 expression, increased ∆K280 tauRD-DsRed solubility, and promoted neurite outgrowth. Thus VB-008 performed best to the end of the present study. The identified compound VB-008 may guide the identification and synthesis of potential inhibitors analogous to this compound.
Collapse
Affiliation(s)
- Chih-Hsin Lin
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University, Taipei 10507, Taiwan
| | - Yu-Shao Hsieh
- Department of Chemistry, National Taiwan Normal University, 88, Ting Chow Road Section 4, Taipei 11677, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University, Taipei 10507, Taiwan
| | - Chia-Jen Hsu
- Department of Chemistry, National Taiwan Normal University, 88, Ting Chow Road Section 4, Taipei 11677, Taiwan
| | - Hsuan-Chiang Chen
- Department of Life Science, National Taiwan Normal University, 88, Ting Chow Road Section 4, Taipei 11677, Taiwan
| | - Wun-Han Huang
- Department of Life Science, National Taiwan Normal University, 88, Ting Chow Road Section 4, Taipei 11677, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University, Taipei 10507, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, 88, Ting Chow Road Section 4, Taipei 11677, Taiwan
| | - Ming-Tsan Su
- Department of Life Science, National Taiwan Normal University, 88, Ting Chow Road Section 4, Taipei 11677, Taiwan
| | - Ying-Chieh Sun
- Department of Chemistry, National Taiwan Normal University, 88, Ting Chow Road Section 4, Taipei 11677, Taiwan.
| | - Guan-Chiun Lee
- Department of Life Science, National Taiwan Normal University, 88, Ting Chow Road Section 4, Taipei 11677, Taiwan.
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, 88, Ting Chow Road Section 4, Taipei 11677, Taiwan.
| |
Collapse
|
37
|
Xiong R, Zhou W, Siegel D, Kitson RRA, Freed CR, Moody CJ, Ross D. A Novel Hsp90 Inhibitor Activates Compensatory Heat Shock Protein Responses and Autophagy and Alleviates Mutant A53T α-Synuclein Toxicity. Mol Pharmacol 2015; 88:1045-54. [PMID: 26405178 PMCID: PMC4658594 DOI: 10.1124/mol.115.101451] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/16/2015] [Indexed: 12/20/2022] Open
Abstract
A potential cause of neurodegenerative diseases, including Parkinson's disease (PD), is protein misfolding and aggregation that in turn leads to neurotoxicity. Targeting Hsp90 is an attractive strategy to halt neurodegenerative diseases, and benzoquinone ansamycin (BQA) Hsp90 inhibitors such as geldanamycin (GA) and 17-(allylamino)-17-demethoxygeldanamycin have been shown to be beneficial in mutant A53T α-synuclein PD models. However, current BQA inhibitors result in off-target toxicities via redox cycling and/or arylation of nucleophiles at the C19 position. We developed novel 19-substituted BQA (19BQA) as a means to prevent arylation. In this study, our data demonstrated that 19-phenyl-GA, a lead 19BQA in the GA series, was redox stable and exhibited little toxicity relative to its parent quinone GA in human dopaminergic SH-SY5Y cells as examined by oxygen consumption, trypan blue, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT), and apoptosis assays. Meanwhile, 19-phenyl-GA retained the ability to induce autophagy and potentially protective heat shock proteins (HSPs) such as Hsp70 and Hsp27. We found that transduction of A53T, but not wild type (WT) α-synuclein, induced toxicity in SH-SY5Y cells. 19-Phenyl-GA decreased oligomer formation and toxicity of A53T α-synuclein in transduced cells. Mechanistic studies indicated that mammalian target of rapamycin (mTOR)/p70 ribosomal S6 kinase signaling was activated by A53T but not WT α-synuclein, and 19-phenyl-GA decreased mTOR activation that may be associated with A53T α-synuclein toxicity. In summary, our results indicate that 19BQAs such as 19-phenyl-GA may provide a means to modulate protein-handling systems including HSPs and autophagy, thereby reducing the aggregation and toxicity of proteins such as mutant A53T α-synuclein.
Collapse
Affiliation(s)
- Rui Xiong
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - Wenbo Zhou
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - Russell R A Kitson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - Curt R Freed
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - Christopher J Moody
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| | - David Ross
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (R.X., D.S., D.R.), and Department of Medicine, Division of Clinical Pharmacology and Toxicology (W.Z., C.R.F.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and School of Chemistry, University of Nottingham, Nottingham, United Kingdom (R.R.A.K., C.J.M.)
| |
Collapse
|
38
|
Braatz EM, Coleman RA. A mathematical model of insulin resistance in Parkinson’s disease. Comput Biol Chem 2015; 56:84-97. [DOI: 10.1016/j.compbiolchem.2015.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 04/06/2015] [Accepted: 04/06/2015] [Indexed: 01/04/2023]
|
39
|
Cox D, Carver JA, Ecroyd H. Preventing α-synuclein aggregation: the role of the small heat-shock molecular chaperone proteins. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1830-43. [PMID: 24973551 DOI: 10.1016/j.bbadis.2014.06.024] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/28/2014] [Accepted: 06/19/2014] [Indexed: 12/21/2022]
Abstract
Protein homeostasis, or proteostasis, is the process of maintaining the conformational and functional integrity of the proteome. The failure of proteostasis can result in the accumulation of non-native proteins leading to their aggregation and deposition in cells and in tissues. The amyloid fibrillar aggregation of the protein α-synuclein into Lewy bodies and Lewy neuritis is associated with neurodegenerative diseases classified as α-synucleinopathies, which include Parkinson's disease and dementia with Lewy bodies. The small heat-shock proteins (sHsps) are molecular chaperones that are one of the cell's first lines of defence against protein aggregation. They act to stabilise partially folded protein intermediates, in an ATP-independent manner, to maintain cellular proteostasis under stress conditions. Thus, the sHsps appear ideally suited to protect against α-synuclein aggregation, yet these fail to do so in the context of the α-synucleinopathies. This review discusses how sHsps interact with α-synuclein to prevent its aggregation and, in doing so, highlights the multi-faceted nature of the mechanisms used by sHsps to prevent the fibrillar aggregation of proteins. It also examines what factors may contribute to α-synuclein escaping the sHsp chaperones in the context of the α-synucleinopathies.
Collapse
Affiliation(s)
- Dezerae Cox
- School of Biological Sciences and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| | - John A Carver
- Research School of Chemistry, The Australian National University, Canberra, Australian Capital Territory, 0200, Australia
| | - Heath Ecroyd
- School of Biological Sciences and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, New South Wales, 2522, Australia.
| |
Collapse
|
40
|
Jones DR, Moussaud S, McLean P. Targeting heat shock proteins to modulate α-synuclein toxicity. Ther Adv Neurol Disord 2014; 7:33-51. [PMID: 24409201 DOI: 10.1177/1756285613493469] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Parkinson's disease is a slowly progressive neurodegenerative disorder typically characterized by the loss of dopaminergic neurons within the substantia nigra pars compacta, and the intraneuronal deposition of insoluble protein aggregates chiefly comprised of α-synuclein. Patients experience debilitating symptoms including bradykinesia, rigidity and postural instability. No curative treatment currently exists and therapeutic strategies are restricted to symptomatic treatment only. Over the past decade a class of molecular chaperones called the heat shock proteins has emerged as a potentially promising therapeutic target. Heat shock proteins aid in the folding and refolding of proteins, and target denatured proteins to degradation systems. By targeting heat shock proteins through various means including overexpression and pharmacological enhancement, researchers have shown that α-synuclein aggregation and its associated cytotoxicity can be therapeutically modulated in an array of cell and animal models. This review highlights the relevant progress in this field and discusses the relevance of heat shock proteins as therapeutic modulators of α-synuclein toxicity to the rapidly evolving understanding of Parkinson's disease pathogenesis.
Collapse
Affiliation(s)
| | | | - Pamela McLean
- Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
41
|
Thakur P, Nehru B. Modulatory effects of sodium salicylate on the factors affecting protein aggregation during rotenone induced Parkinson's disease pathology. Neurochem Int 2014; 75:1-10. [PMID: 24852355 DOI: 10.1016/j.neuint.2014.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/06/2014] [Accepted: 05/09/2014] [Indexed: 01/20/2023]
Abstract
Sodium salicylate (SS) confers neuroprotection in various models of Parkinson's disease (PD) but the mechanisms behind its protective actions are not clear. PD pathology is multifactorial involving numerous processes such as protein aggregation, dysfunction of protein degradation machinery and apoptosis. Detailed evaluation of effects of SS on these processes can provide an insight into the mechanism of neuroprotection by SS in PD pathology. In a rotenone (2mg/kg b.w.) based rat model of PD, SS (100mg/kg b.w.) was administered in conjunction. Drug treatments continued for 5 weeks after which various analyses were conducted using mid-brain tissue. IHC analysis revealed a decline in the aggregation of α-synuclein and ubiquitin with SS supplementation. These effects might be mediated by the elevation in HSF-1, HSP-40, and HSP-27 expression following SS co-treatment. This HSP upregulation helped in the improvement in proteasome activity as well as expression. Further, IHC analysis revealed that SS co-treatment prevented the activation of astrocytes caused by rotenone. Since astrocytes are involved in maintenance of glutathione (GSH) homeostasis, it resulted in a concomitant improvement in the GSH levels. As a result, decrease in apoptosis as indicated by caspase-9 and caspase-3 expression as well as TUNEL assay was also observed in the SS conjunction group. Our results indicate that besides being a known free radical scavenger and anti-inflammatory compound, SS can provide neuroprotection by differently upregulating the HSPs and reducing the protein aggregation burden.
Collapse
Affiliation(s)
- Poonam Thakur
- Department of Biophysics, Panjab University, Chandigarh 160014, India.
| | - Bimla Nehru
- Department of Biophysics, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
42
|
Gao H, Liu X, Chen D, Lv L, Wu M, Mi J, Wang W. Comparative study of Hsp27, GSK3β, Wnt1 and PRDX3 in Hirschsprung's disease. Int J Exp Pathol 2014; 95:229-37. [PMID: 24773279 DOI: 10.1111/iep.12075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 01/29/2014] [Indexed: 01/20/2023] Open
Abstract
Hirschsprung's disease (HSCR) is a developmental disorder of the enteric nervous system characterized by aganglionosis in distal gut. In this study, we used two-dimensional gel electrophoresis (2-DE) technology coupled with matrix assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) analysis to identify differentially expressed proteins in the aganglionic (stenotic) and ganglionic (normal) colon segment tissues from patients with HSCR. We identified 15 proteins with different expression levels between the stenotic and the normal colon segment tissues from patients with HSCR. Nine proteins were upregulated and six proteins downregulated in the stenotic colon segment tissues compared to the normal colon segment tissues. Based on the biological functions, we selected the Hsp27 upregulated proteins and the PRDX3 downregulated proteins to confirm their expression in 20 patients. The protein and mRNA expressions of Hsp27 were statistically higher in the stenotic colon segment tissues than in the normal colon segment tissues, whereas the protein and mRNA expressions of PRDX3 were statistically lower in the stenotic colon segment tissues than in the normal colon segment tissues. These findings of changes in mRNA and protein in tissues from patients with HSCR provide information which may be helpful in understanding the pathomechanism that is implicated in the disease.
Collapse
Affiliation(s)
- Hong Gao
- Laboratory of Pediatric Congenital Malformation, Ministry of Public Health, Shengjing Hospital of China Medical University, Shenyang, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Thakur P, Nehru B. Long-term heat shock proteins (HSPs) induction by carbenoxolone improves hallmark features of Parkinson's disease in a rotenone-based model. Neuropharmacology 2014; 79:190-200. [DOI: 10.1016/j.neuropharm.2013.11.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/10/2013] [Accepted: 11/20/2013] [Indexed: 01/24/2023]
|
44
|
Mode of action of S-methyl-N, N-diethylthiocarbamate sulfoxide (DETC-MeSO) as a novel therapy for stroke in a rat model. Mol Neurobiol 2014; 50:655-72. [PMID: 24573692 DOI: 10.1007/s12035-014-8658-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/05/2014] [Indexed: 10/25/2022]
Abstract
One approach for protecting neurons from excitotoxic damage in stroke is to attenuate receptor activity with specific antagonists. S-Methyl-N, N-diethylthiocarbamate sulfoxide (DETC-MeSO), the active metabolite of disulfiram, has been shown to be a partial antagonist of glutamate receptors and effective in reducing seizure. First, we investigated neuroprotective effect of DETC-MeSO on primary cortical neuronal culture under hypoxia/reoxygenation condition in vitro. Then, DETC-MeSO was administered subcutaneously for 4 and 8 days with the first injection occurring 1 h before or 24 h after reperfusion in the rat middle cerebral artery occlusion stroke model. Rats were subjected to the neuroscore test, and the brain was analyzed for infarct size. Monitoring neurotransmitter release was carried out by microdialysis. Heat shock proteins, key proteins involved in apoptosis and endoplasmic reticulum (ER) stress, were analyzed by immunoblotting. DETC-MeSO greatly reduced both cell death following hypoxia/reoxygenation and brain infarct size. It improved performance on the neuroscore test and attenuated proteolysis of αII-spectrin. The level of pro-apoptotic proteins declined, and anti-apoptotic and HSP27 protein expressions were markedly increased. Levels of the ER stress protein markers p-PERK, p-eIF2α, ATF4, JNK, XBP-1, GADD34, and CHOP significantly declined after DETC-MeSO administration. Microdialysis data showed that DETC-MeSO increased high potassium-induced striatal dopamine release indicating that more neurons were protected and survived under ischemic insult in the presence of DETC-MeSO. We also showed that DETC-MeSO can prevent gliosis. DETC-MeSO elicits neuroprotection through the preservation of ER resulting in reduction of apoptosis by increase of anti-apoptotic proteins and decrease of pro-apoptotic proteins.
Collapse
|
45
|
Ebrahimi-Fakhari D, Saidi LJ, Wahlster L. Molecular chaperones and protein folding as therapeutic targets in Parkinson's disease and other synucleinopathies. Acta Neuropathol Commun 2013; 1:79. [PMID: 24314025 PMCID: PMC4046681 DOI: 10.1186/2051-5960-1-79] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/25/2013] [Indexed: 12/20/2022] Open
Abstract
Changes in protein metabolism are key to disease onset and progression in many neurodegenerative diseases. As a prime example, in Parkinson's disease, folding, post-translational modification and recycling of the synaptic protein α-synuclein are clearly altered, leading to a progressive accumulation of pathogenic protein species and the formation of intracellular inclusion bodies. Altered protein folding is one of the first steps of an increasingly understood cascade in which α-synuclein forms complex oligomers and finally distinct protein aggregates, termed Lewy bodies and Lewy neurites. In neurons, an elaborated network of chaperone and co-chaperone proteins is instrumental in mediating protein folding and re-folding. In addition to their direct influence on client proteins, chaperones interact with protein degradation pathways such as the ubiquitin-proteasome-system or autophagy in order to ensure the effective removal of irreversibly misfolded and potentially pathogenic proteins. Because of the vital role of proper protein folding for protein homeostasis, a growing number of studies have evaluated the contribution of chaperone proteins to neurodegeneration. We herein review our current understanding of the involvement of chaperones, co-chaperones and chaperone-mediated autophagy in synucleinopathies with a focus on the Hsp90 and Hsp70 chaperone system. We discuss genetic and pathological studies in Parkinson's disease as well as experimental studies in models of synucleinopathies that explore molecular chaperones and protein degradation pathways as a novel therapeutic target. To this end, we examine the capacity of chaperones to prevent or modulate neurodegeneration and summarize the current progress in models of Parkinson's disease and related neurodegenerative disorders.
Collapse
|
46
|
Moloney TC, Hyland R, O'Toole D, Paucard A, Kirik D, O'Doherty A, Gorman AM, Dowd E. Heat shock protein 70 reduces α-synuclein-induced predegenerative neuronal dystrophy in the α-synuclein viral gene transfer rat model of Parkinson's disease. CNS Neurosci Ther 2013; 20:50-8. [PMID: 24279716 DOI: 10.1111/cns.12200] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 10/02/2013] [Accepted: 10/06/2013] [Indexed: 01/20/2023] Open
Abstract
AIMS It has become increasingly evident that the nigrostriatal degeneration associated with Parkinson's disease initiates at the level of the axonal terminals in the putamen, and this nigrostriatal terminal dystrophy is either caused or exacerbated by the presence of α-synuclein immunopositive neuronal inclusions. Therefore, strategies aimed at reducing α-synuclein-induced early neuronal dystrophy may slow or halt the progression to overt nigrostriatal neurodegeneration. Thus, this study sought to determine if adeno-associated virus (AAV) mediated overexpression of two molecular chaperone heat shock proteins, namely Hsp27 or Hsp70, in the AAV-α-synuclein viral gene transfer rat model of Parkinson's disease could prevent α-synuclein-induced early neuronal pathology. METHODS Male Sprague-Dawley rats were intranigrally coinjected with pathogenic (AAV-α-synuclein) and putative therapeutic (AAV-Hsp27 or AAV-Hsp70) viral vectors and were sacrificed 18 weeks postviral injection. RESULTS Intranigral injection of AAV-α-synuclein resulted in significant α-synuclein accumulation in the substantia nigra and striatal terminals which led to significant dystrophy of nigrostriatal dopaminergic neurons without overt nigrostriatal neurodegeneration. Coinjection of AAV-Hsp70, but not AAV-Hsp27, significantly reduced AAV-α-synuclein-induced neuronal dystrophy. CONCLUSIONS These data confirm that overexpression of Hsp70 holds significant potential as a disease-modulating therapeutic approach for Parkinson's disease, with protective effects against early-onset α-synuclein-induced pathology demonstrated in the AAV-α-synuclein model.
Collapse
Affiliation(s)
- Teresa C Moloney
- Department of Pharmacology & Therapeutics, National University of Ireland, Galway, Ireland; National Centre for Biomedical Engineering Science (NCBES), National University of Ireland, Galway, Ireland; NCBES Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Kilpatrick K, Novoa JA, Hancock T, Guerriero CJ, Wipf P, Brodsky JL, Segatori L. Chemical induction of Hsp70 reduces α-synuclein aggregation in neuroglioma cells. ACS Chem Biol 2013; 8:1460-8. [PMID: 23594135 DOI: 10.1021/cb400017h] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Misfolding and aggregation of α-synuclein (α-syn) is associated with the development of a number of neurodegenerative diseases including Parkinson's disease (PD). Analyses of post mortem tissues revealed the presence of molecular chaperones within α-syn aggregates, suggesting that chaperones play a role in α-syn misfolding and aggregation. In fact, inhibition of chaperone activity aggravates α-syn toxicity, and the overexpression of chaperones, particularly 70-kDa heat shock protein (Hsp70), protects against α-syn-induced toxicity. In this study, we investigated the effect of carbenoxolone (CBX), a glycyrrhizic acid derivative previously reported to upregulate Hsp70, in human neuroglioma cells overexpressing α-syn. We report that CBX treatment lowers α-syn aggregation and prevents α-syn-induced cytotoxicity. We demonstrate further that Hsp70 induction by CBX arises from activation of heat shock factor 1 (HSF1). The Hsp70 inhibitor MAL3-101 and the Hsp70 enhancer 115-7c led to an increase or decrease in α-syn aggregation, respectively, in agreement with these findings. In summary, this study provides a proof-of-principle demonstration that chemical modulation of the Hsp70 machine is a promising strategy to prevent α-syn aggregation.
Collapse
Affiliation(s)
- Kiri Kilpatrick
- Departments of †Chemical
and Biomolecular Engineering, ∥Bioengineering, and ⊥Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
- Departments of §Chemistry and ‡Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
15260, United States
| | - Jose Andres Novoa
- Departments of †Chemical
and Biomolecular Engineering, ∥Bioengineering, and ⊥Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
- Departments of §Chemistry and ‡Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
15260, United States
| | - Tommy Hancock
- Departments of †Chemical
and Biomolecular Engineering, ∥Bioengineering, and ⊥Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
- Departments of §Chemistry and ‡Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
15260, United States
| | - Christopher J. Guerriero
- Departments of †Chemical
and Biomolecular Engineering, ∥Bioengineering, and ⊥Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
- Departments of §Chemistry and ‡Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
15260, United States
| | - Peter Wipf
- Departments of †Chemical
and Biomolecular Engineering, ∥Bioengineering, and ⊥Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
- Departments of §Chemistry and ‡Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
15260, United States
| | - Jeffrey L. Brodsky
- Departments of †Chemical
and Biomolecular Engineering, ∥Bioengineering, and ⊥Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
- Departments of §Chemistry and ‡Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
15260, United States
| | - Laura Segatori
- Departments of †Chemical
and Biomolecular Engineering, ∥Bioengineering, and ⊥Biochemistry and Cell Biology, Rice University, Houston, Texas 77005,
United States
- Departments of §Chemistry and ‡Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
15260, United States
| |
Collapse
|
48
|
van der Putten H, Lotz GP. Opportunities and challenges for molecular chaperone modulation to treat protein-conformational brain diseases. Neurotherapeutics 2013; 10:416-28. [PMID: 23536253 PMCID: PMC3701765 DOI: 10.1007/s13311-013-0186-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A common pathological hallmark of protein-conformational brain diseases is the formation of disease-specific protein aggregates. In Alzheimer's disease, these are comprised of amyloid-β and Tau as opposed to α-synuclein in Parkinson's disease and N-terminal fragments of mutant huntingtin in Huntington's disease. Most aggregates also sequester molecular chaperones, a protein family that assists in the folding, refolding, stabilization, and processing of client proteins, including misfolded proteins in brain diseases. Molecular chaperone modulation has achieved remarkable therapeutic effects in some cellular and preclinical animal models of protein-conformational diseases. This has raised hope for chaperone-based strategies to combat these diseases. Here, we review briefly the functional diversity and medical significance of molecular chaperones, their therapeutic potential, and common and specific challenges towards clinical application.
Collapse
Affiliation(s)
- Herman van der Putten
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland
| | - Gregor P. Lotz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland
| |
Collapse
|
49
|
Chaari A, Hoarau-Véchot J, Ladjimi M. Applying chaperones to protein-misfolding disorders: molecular chaperones against α-synuclein in Parkinson's disease. Int J Biol Macromol 2013; 60:196-205. [PMID: 23748003 DOI: 10.1016/j.ijbiomac.2013.05.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 05/28/2013] [Accepted: 05/29/2013] [Indexed: 02/04/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the accumulation of a protein called α-synuclein (α-syn) into inclusions known as lewy bodies (LB) within neurons. This accumulation is also due to insufficient formation and activity of dopamine produced in certain neurons within the substantia nigra. Lewy bodies are the pathological hallmark of the idiopathic disorder and the cascade that allows α-synuclein to misfold, aggregate and form these inclusions has been the subject of intensive research. Targeting these early steps of oligomerization is one of the main therapeutic approaches in order to develop neurodegenerative-modifying agents. Because the folding and refolding of alpha synuclein is the key point of this cascade, we are interested in this review to summarize the role of some molecular chaperones proteins such as Hsp70, Hsp90 and small heat shock proteins (sHsp) and Hsp 104. Hsp70 and its co-chaperone, Hsp70 and small heat shock proteins can prevent neurodegeneration by preventing α-syn misfolding, oligomerization and aggregation in vitro and in Parkinson disease animal models. Hsp104 is able to resolve disordered protein aggregates and cross beta amyloid conformers. Together, these chaperones have a complementary effect and can be a target for therapeutic intervention in PD.
Collapse
Affiliation(s)
- Ali Chaari
- Department of Biochemistry, Weill Cornell Medical College in Qatar, Qatar Foundation, Education City, Doha, Qatar.
| | | | | |
Collapse
|
50
|
Sabirzhanov B, Stoica BA, Hanscom M, Piao CS, Faden AI. Over-expression of HSP70 attenuates caspase-dependent and caspase-independent pathways and inhibits neuronal apoptosis. J Neurochem 2012; 123:542-54. [PMID: 22909049 DOI: 10.1111/j.1471-4159.2012.07927.x] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 08/13/2012] [Accepted: 08/14/2012] [Indexed: 01/22/2023]
Abstract
HSP70 is a member of the family of heat-shock proteins that are known to be up-regulated in neurons following injury and/or stress. HSP70 over-expression has been linked to neuroprotection in multiple models, including neurodegenerative disorders. In contrast, less is known about the neuroprotective effects of HSP70 in neuronal apoptosis and with regard to modulation of programmed cell death (PCD) mechanisms in neurons. We examined the effects of HSP70 over-expression by transfection with HSP70-expression plasmids in primary cortical neurons and the SH-SY5Y neuronal cell line using four independent models of apoptosis: etoposide, staurosporine, C2-ceramide, and β-Amyloid. In these apoptotic models, neurons transfected with the HSP70 construct showed significantly reduced induction of nuclear apoptotic markers and/or cell death. Furthermore, we demonstrated that HSP70 binds and potentially inactivates Apoptotic protease-activating factor 1, as well as apoptosis-inducing factor, key molecules involved in development of caspase-dependent and caspase-independent PCD, respectively. Markers of caspase-dependent PCD, including active caspase-3, caspase-9, and cleaved PARP were attenuated in neurons over-expressing HSP70. These data indicate that HSP70 protects against neuronal apoptosis and suggest that these effects reflect, at least in part, to inhibition of both caspase-dependent and caspase-independent PCD pathways.
Collapse
Affiliation(s)
- Boris Sabirzhanov
- Department of Anesthesiology, Shock Trauma & Anesthesiology Research (STAR) Organized Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | | | | | |
Collapse
|