1
|
Chierichetti M, Cristofani R, Crippa V, Ferrari V, Cozzi M, Casarotto E, Pramaggiore P, Cornaggia L, Patelli G, Mohamed A, Piccolella M, Galbiati M, Rusmini P, Tedesco B, Poletti A. Small heat shock protein B8: from cell functions to its involvement in diseases and potential therapeutic applications. Neural Regen Res 2025; 20:2872-2886. [PMID: 39435632 PMCID: PMC11826450 DOI: 10.4103/nrr.nrr-d-24-00517] [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: 05/06/2024] [Revised: 08/05/2024] [Accepted: 08/29/2024] [Indexed: 10/23/2024] Open
Abstract
Heat shock protein family B (small) member 8 (HSPB8) is a 22 kDa ubiquitously expressed protein belonging to the family of small heat shock proteins. HSPB8 is involved in various cellular mechanisms mainly related to proteotoxic stress response and in other processes such as inflammation, cell division, and migration. HSPB8 binds misfolded clients to prevent their aggregation by assisting protein refolding or degradation through chaperone-assisted selective autophagy. In line with this function, the pro-degradative activity of HSPB8 has been found protective in several neurodegenerative and neuromuscular diseases characterized by protein misfolding and aggregation. In cancer, HSPB8 has a dual role being capable of exerting either a pro- or an anti-tumoral activity depending on the pathways and factors expressed by the model of cancer under investigation. Moreover, HSPB8 exerts a protective function in different diseases by modulating the inflammatory response, which characterizes not only neurodegenerative diseases, but also other chronic or acute conditions affecting the nervous system, such as multiple sclerosis and intracerebellar hemorrhage. Of note, HSPB8 modulation may represent a therapeutic approach in other neurological conditions that develop as a secondary consequence of other diseases. This is the case of cognitive impairment related to diabetes mellitus, in which HSPB8 exerts a protective activity by assuring mitochondrial homeostasis. This review aims to summarize the diverse and multiple functions of HSPB8 in different pathological conditions, focusing on the beneficial effects of its modulation. Drug-based and alternative therapeutic approaches targeting HSPB8 and its regulated pathways will be discussed, emphasizing how new strategies for cell and tissue-specific delivery represent an avenue to advance in disease treatments.
Collapse
Affiliation(s)
- Marta Chierichetti
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Riccardo Cristofani
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Valeria Crippa
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Veronica Ferrari
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Marta Cozzi
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Elena Casarotto
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Paola Pramaggiore
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Laura Cornaggia
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Guglielmo Patelli
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Ali Mohamed
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Margherita Piccolella
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Mariarita Galbiati
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Paola Rusmini
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Barbara Tedesco
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Angelo Poletti
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
2
|
Das S, Rajeswari VD, Venkatraman G, Ramanathan G. Phytochemicals in Parkinson's Disease: a Pathway to Neuroprotection and Personalized Medicine. Cell Biochem Biophys 2025; 83:1427-1443. [PMID: 39537915 DOI: 10.1007/s12013-024-01607-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder marked by the progressive loss of dopaminergic neurons in the substantia nigra. While current treatments primarily manage symptoms, there is increasing interest in alternative approaches, particularly the use of phytochemicals from medicinal plants. These natural compounds have demonstrated promising neuroprotective potential in preclinical studies by targeting key pathological mechanisms such as oxidative stress, neuroinflammation, and protein aggregation. However, the clinical translation of these phytochemicals is limited due to a lack of robust clinical trials evaluating their safety, efficacy, and pharmacokinetics. This review provides a comprehensive overview of the neuroprotective potential of phytochemicals in PD management, examining the mechanisms underlying PD pathogenesis and emphasizing neuroprotection. It explores the historical and current research on medicinal plants like Mucuna pruriens, Curcuma longa, and Ginkgo biloba, and discusses the challenges in clinical translation, including ethical and practical considerations and the integration with conventional therapies. It further underscores the need for future research to elucidate mechanisms of action, optimize drug delivery, and conduct rigorous clinical trials to establish the safety and efficacy of phytochemicals, aiming to shape future neuroprotective strategies and develop more effective, personalized treatments for PD.
Collapse
Affiliation(s)
- Soumik Das
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - V Devi Rajeswari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Ganesh Venkatraman
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Gnanasambandan Ramanathan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, India.
| |
Collapse
|
3
|
HEALEY ALS Platform Trial, HEALEY ALS Platform Trial Study Group. Safety and efficacy of trehalose in amyotrophic lateral sclerosis (HEALEY ALS Platform Trial): an adaptive, phase 2/3, double-blind, randomised, placebo-controlled trial. Lancet Neurol 2025; 24:500-511. [PMID: 40409314 DOI: 10.1016/s1474-4422(25)00173-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/28/2025] [Accepted: 05/02/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND Trehalose is a disaccharide that activates autophagy pathways in animal models of neurodegenerative diseases, with the potential to catalyse clearance of toxic, misfolded proteins in motor neurons and slow disease progression in amyotrophic lateral sclerosis (ALS). We aimed to evaluate the safety and efficacy of trehalose in individuals with ALS. METHODS The HEALEY ALS Platform Trial is a perpetual, adaptive, phase 2/3, randomised, double-blind, multi-regimen trial conducted at 60 geographically diverse sites in the USA. In the current regimen, adults with clinically possible, probable, laboratory-supported probable, or definite ALS, defined by the revised El Escorial criteria, were randomly allocated (3:1), stratified by use of edaravone and riluzole, to receive trehalose 0·75 g per kg intravenously weekly over 24 weeks, or matching placebo. The primary outcome was a composite of the relative rate of disease progression, as measured by the Revised ALS Functional Rating Scale (ALSFRS-R), and survival over 24 weeks, estimated in a Bayesian shared-parameter model. The study included prespecified stopping rules for futility; interim analyses occurred every 12 weeks. The primary outcome was analysed according to the intention-to-treat principle in all participants in the trehalose group, the placebo group within the regimen, and placebo groups from other contributing regimens; the safety analysis population was comprised of all participants who initiated treatment. This study is registered with ClinicalTrials.gov, NCT05136885. FINDINGS Between Feb 21, 2022, and Feb 17, 2023, 1021 participants were screened for the platform trial and 171 were assigned to the trehalose regimen. Of these, 161 participants met eligibility criteria, with 120 randomly allocated to trehalose and 41 to regimen-specific placebo. 164 participants randomly allocated to placebo in other regimens were added for analysis (totalling 205 placebo recipients). The disease rate ratio for change in ALSFRS-R and survival was 0·87 (95% credible interval 0·665-1·102, posterior probability of superiority 0·877). Serious adverse events occurred in 19 (16%) participants in the trehalose group and three (7%) participants in the regimen-only placebo group, leading to premature discontinuations in 14 (12%) versus one (2%), respectively. Fatal treatment-emergent adverse events occurred in seven participants in the trehalose group and none in the regimen-only placebo group. No death was considered related to the trial drug. The most common cause of death was respiratory failure, consistent with the natural history of ALS. INTERPRETATION Trehalose was well tolerated but there was no evidence to suggest a difference in ALS disease progression compared with placebo in this study. No statistical benefit was seen in secondary clinical or biomarker measures, suggesting that trehalose at this dosage is unlikely to be efficacious for treatment of ALS. FUNDING AMG Charitable Foundation, Tackle ALS, the ALS Association, ALS Finding a Cure, the Muscular Dystrophy Association, ALS ONE, the Arthur M Blank Family Foundation, I AM ALS, Tambourine ALS Collaborative, and other community fundraising initiatives and donors. Study drug and partial regimen-related funding was provided by Seelos.
Collapse
|
4
|
Maruf A, Gerasymchuk D, Hlushchuk I, Er S, Milewska M, Chmielarz P, Domanskyi A, Airavaara M, Wandzik I. Trehalose-releasing nanogels reduce α-synuclein-induced Lewy body-like inclusions in primary mouse hippocampal neurons. J Mater Chem B 2025; 13:5845-5857. [PMID: 40289859 DOI: 10.1039/d4tb02704c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Parkinson's disease (PD) is the second most prevalent age-related neurodegenerative disorder, clinically characterized by both motor and non-motor symptoms. A key hallmark of PD is the accumulation of misfolded α-synuclein, which aggregates to Lewy bodies (LB) formed inside neurons. Trehalose, a disaccharide that induces autophagy, has been demonstrated to reduce α-synuclein aggregation in vivo. However, the enzyme trehalase rapidly degrades free trehalose, and its hydrophilicity causes poor penetration through the cell membrane. Thus, advanced trehalose delivery strategies are urgently needed. Herein, we investigated the effects of trehalose-bearing nanogels for reducing α-synuclein protein-induced perinuclear LB-like pathology in primary mouse hippocampal neurons. The study compares the effects of trehalose-releasing nanogel (TR) and trehalose-non releasing nanogel (TNR). The results showed that TR, but not TNR, nor free trehalose reduced LB-like inclusions in primary hippocampal neurons. The neuroprotective effects of TR may result from the synergistic effects of direct limitation of α-synuclein aggregates formation and trehalose release-induced autophagy promoting aggregates clearance. Overall, enhancing trehalose delivery with nanogels that can sustainably release trehalose could be worth further investigation as a new potential option for reducing α-synuclein aggregation in neurons affected by neurodegenerative diseases.
Collapse
Affiliation(s)
- Ali Maruf
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland.
- Biotechnology Center, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland
- Division of Pharmacology and Pharmacotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland.
| | - Dmytro Gerasymchuk
- Division of Pharmacology and Pharmacotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland.
| | - Irena Hlushchuk
- Division of Pharmacology and Pharmacotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland.
| | - Safak Er
- Division of Pharmacology and Pharmacotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland.
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Małgorzata Milewska
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland.
- Biotechnology Center, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland
| | - Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki 00014, Finland
| | - Mikko Airavaara
- Division of Pharmacology and Pharmacotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland.
| | - Ilona Wandzik
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego 4, 44-100 Gliwice, Poland.
- Biotechnology Center, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland
| |
Collapse
|
5
|
Ramos-Molina AR, Tejeda-Martínez AR, Viveros-Paredes JM, Chaparro-Huerta V, Urmeneta-Ortíz MF, Ramírez-Jirano LJ, Flores-Soto ME. Beta-caryophyllene inhibits the permeability of the blood-brain barrier in MPTP-induced parkinsonism. Neurologia 2025; 40:191-203. [PMID: 40054982 DOI: 10.1016/j.nrleng.2025.02.003] [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: 03/18/2022] [Accepted: 11/29/2022] [Indexed: 05/13/2025] Open
Abstract
INTRODUCTION Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder worldwide. Although the precise pathogenesis of PD remains unclear, several studies demonstrate that oxidative stress, inflammation, low levels of antioxidants, and the presence of biomolecules that generate reactive oxygen species can disrupt the blood-brain barrier (BBB) as an essential feature of the disease. AIMS This study aimed to test whether agonism to cannabinoid receptor type 2 (CB2) through the administration of β-caryophyllene (BCP) could correct BBB permeability in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) parkinsonism induction model. METHODS We conducted a molecular assessment of proteins (immunochemistry and western blot), BBB permeability, and related biomarkers of PD (lipid peroxidation) in the MPTP mouse model of the disease. RESULTS Expression of zonula occludens (ZO-1) and occludin tight junction (TJ) proteins was dampened in the striatum and substantia nigra pars compacta of mice, while lipid peroxidation and BBB permeability increased in the striatum in the MPTP-treated group, and these effects were reversed under BCP administration. This phytocannabinoid was able to restore protein expression and immunoreactivity of tyrosine hydroxylase (TH), ionized calcium-binding adapter molecule 1 (Iba-1), and glial fibrillary acidic protein (GFAP), as well as nuclear factor-erythroid 2-related factor (NRF2) translocation to the nucleus, and NADPH quinone oxidase 1 (NQO1) expression in mice treated with MPTP. CONCLUSION These results highlight the role of CB2 as a therapeutic target for PD, suggesting that its activation may ameliorate PD-related BBB disruption and oxidative stress, reducing the selective death of dopaminergic neurons.
Collapse
Affiliation(s)
- A R Ramos-Molina
- Centro Universitario de los Lagos, Universidad de Guadalajara, Lagos de Moreno, Jalisco, Mexico; Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - A R Tejeda-Martínez
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - J M Viveros-Paredes
- Laboratorio de Investigación y Desarrollo Farmacéutico, Departamento de Farmacología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - V Chaparro-Huerta
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico
| | - M F Urmeneta-Ortíz
- Centro Universitario de los Lagos, Universidad de Guadalajara, Lagos de Moreno, Jalisco, Mexico
| | - L J Ramírez-Jirano
- División de Neurociencias, Centro de Investigación Biomédica Occidente (IMSS), Guadalajara, Mexico
| | - M E Flores-Soto
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
6
|
Pradeloux S, Coulombe K, Ouamba AJK, Isenbrandt A, Calon F, Roy D, Soulet D. Oral Trehalose Intake Modulates the Microbiota-Gut-Brain Axis and Is Neuroprotective in a Synucleinopathy Mouse Model. Nutrients 2024; 16:3309. [PMID: 39408276 PMCID: PMC11478413 DOI: 10.3390/nu16193309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/03/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease affecting dopaminergic neurons in the nigrostriatal and gastrointestinal tracts, causing both motor and non-motor symptoms. This study examined the neuroprotective effects of trehalose. This sugar is confined in the gut due to the absence of transporters, so we hypothesized that trehalose might exert neuroprotective effects on PD through its action on the gut microbiota. We used a transgenic mouse model of PD (PrP-A53T G2-3) overexpressing human α-synuclein and developing GI dysfunctions. Mice were given water with trehalose, maltose, or sucrose (2% w/v) for 6.5 m. Trehalose administration prevented a reduction in tyrosine hydroxylase immunoreactivity in the substantia nigra (-25%), striatum (-38%), and gut (-18%) in PrP-A53T mice. It also modulated the gut microbiota, reducing the loss of diversity seen in PrP-A53T mice and promoting bacteria negatively correlated with PD in patients. Additionally, trehalose treatment increased the intestinal secretion of glucagon-like peptide 1 (GLP-1) by 29%. Maltose and sucrose, which break down into glucose, did not show neuroprotective effects, suggesting glucose is not involved in trehalose-mediated neuroprotection. Since trehalose is unlikely to cross the intestinal barrier at the given dose, the results suggest its effects are mediated indirectly through the gut microbiota and GLP-1.
Collapse
Affiliation(s)
- Solène Pradeloux
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Katherine Coulombe
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Alexandre Jules Kennang Ouamba
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté des Sciences de l’Agriculture et de l’Alimentation, Université Laval, Québec, QC G1V 0A6, Canada
| | - Amandine Isenbrandt
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Frédéric Calon
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Denis Roy
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté des Sciences de l’Agriculture et de l’Alimentation, Université Laval, Québec, QC G1V 0A6, Canada
| | - Denis Soulet
- Centre de Recherche du CHU de Québec, Québec, QC G1V 4G2, Canada; (S.P.); (K.C.); (A.J.K.O.); (A.I.); (F.C.); (D.R.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
7
|
Tanaka MT, Miki Y, Mori F, Kon T, Furukawa T, Shimoyama S, Tatara Y, Ozaki T, Bettencourt C, Warner TT, Wakabayashi K. Intranasal administration of trehalose reduces α-synuclein oligomers and accelerates α-synuclein aggregation. Brain Commun 2024; 6:fcae193. [PMID: 39165481 PMCID: PMC11334933 DOI: 10.1093/braincomms/fcae193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/08/2024] [Accepted: 06/06/2024] [Indexed: 08/22/2024] Open
Abstract
Abnormal α-synuclein (αSyn), including an oligomeric form of αSyn, accumulates and causes neuronal dysfunction in the brains of patients with multiple system atrophy. Neuroprotective drugs that target abnormal αSyn aggregation have not been developed for the treatment of multiple system atrophy. In addition, treating diseases at an early stage is crucial to halting the progress of neuronal damage in neurodegeneration. In this study, using early-stage multiple system atrophy mouse model and in vitro kinetic analysis, we investigated how intranasal and oral administration of trehalose can improve multiple system atrophy pathology and clinical symptoms. The multiple system atrophy model showed memory impairment at least four weeks after αSyn induction. Behavioural and physiological analyses showed that intranasal and oral administration of trehalose reversed memory impairments to near-normal levels. Notably, trehalose treatment reduced the amount of toxic αSyn and increased the aggregated form of αSyn in the multiple system atrophy model brain. In vitro kinetic analysis confirmed that trehalose accelerated the aggregate formation of αSyn. Based on our findings, we propose a novel strategy whereby accelerated αSyn aggregate formation leads to reduced exposure to toxic αSyn oligomers, particularly during the early phase of disease progression.
Collapse
Affiliation(s)
- Makoto T Tanaka
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Yasuo Miki
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| | - Fumiaki Mori
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Tomoya Kon
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Tomonori Furukawa
- Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Shuji Shimoyama
- Department of Neurophysiology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Yota Tatara
- Department of Stress Response Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Taku Ozaki
- Department of Biological Science, Graduate School of Science and Engineering, Iwate University, Morioka 020-8551, Japan
| | - Conceição Bettencourt
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Thomas T Warner
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Koichi Wakabayashi
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| |
Collapse
|
8
|
Bejarano E, Domenech-Bendaña A, Avila-Portillo N, Rowan S, Edirisinghe S, Taylor A. Glycative stress as a cause of macular degeneration. Prog Retin Eye Res 2024; 101:101260. [PMID: 38521386 PMCID: PMC11699537 DOI: 10.1016/j.preteyeres.2024.101260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
People are living longer and rates of age-related diseases such as age-related macular degeneration (AMD) are accelerating, placing enormous burdens on patients and health care systems. The quality of carbohydrate foods consumed by an individual impacts health. The glycemic index (GI) is a kinetic measure of the rate at which glucose arrives in the blood stream after consuming various carbohydrates. Consuming diets that favor slowly digested carbohydrates releases sugar into the bloodstream gradually after consuming a meal (low glycemic index). This is associated with reduced risk for major age-related diseases including AMD, cardiovascular disease, and diabetes. In comparison, consuming the same amounts of different carbohydrates in higher GI diets, releases glucose into the blood rapidly, causing glycative stress as well as accumulation of advanced glycation end products (AGEs). Such AGEs are cytotoxic by virtue of their forming abnormal proteins and protein aggregates, as well as inhibiting proteolytic and other protective pathways that might otherwise selectively recognize and remove toxic species. Using in vitro and animal models of glycative stress, we observed that consuming higher GI diets perturbs metabolism and the microbiome, resulting in a shift to more lipid-rich metabolomic profiles. Interactions between aging, diet, eye phenotypes and physiology were observed. A large body of laboratory animal and human clinical epidemiologic data indicates that consuming lower GI diets, or lower glycemia diets, is protective against features of early AMD (AMDf) in mice and AMD prevalence or AMD progression in humans. Drugs may be optimized to diminish the ravages of higher glycemic diets. Human trials are indicated to determine if AMD progression can be retarded using lower GI diets. Here we summarized the current knowledge regarding the pathological role of glycative stress in retinal dysfunction and how dietary strategies might diminish retinal disease.
Collapse
Affiliation(s)
- Eloy Bejarano
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Alicia Domenech-Bendaña
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | | | - Sheldon Rowan
- JM USDA Human Nutrition Research Center on Aging at Tufts University, United States
| | - Sachini Edirisinghe
- Tufts University Friedman School of Nutrition Science and Policy, United States
| | - Allen Taylor
- Tufts University Friedman School of Nutrition Science and Policy, United States.
| |
Collapse
|
9
|
Zhu D, Zhang S, Wang X, Xiao C, Cui G, Yang X. Secretory Clusterin Inhibits Dopamine Neuron Apoptosis in MPTP Mice by Preserving Autophagy Activity. Neuroscience 2024; 540:38-47. [PMID: 38242280 DOI: 10.1016/j.neuroscience.2024.01.010] [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: 03/01/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Secretory clusterin (sCLU) plays an important role in the research progress of nervous system diseases. However, the physiological function of sCLU in Parkinson's disease (PD) are unclear. The purpose of this study was to examine the effects of sCLU-mediated autophagy on cell survival and apoptosis inhibition in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. We found that MPTP administration induced prolonged pole-climbing time, shortened traction time and rotarod time, significantly decreased TH protein expression in the SN tissue of mice. In contrast, sCLU -treated mice took less time to climb the pole and had an extended traction time and rotating rod time. Meanwhile, sCLU intervention induced increased expression of the TH protein in the SN of mice. These results indicated that sCLU intervention could reduce the loss of dopamine neurons in the SN area and alleviate dyskinesia in mice. Furthermore, MPTP led to suppressed viability, enhanced apoptosis, an increased Bax/Bcl-2 ratio, and cleaved caspase-3 in the SN of mice, and these effects were abrogated by sCLU intervention. In addition, MPTP increased the levels of P62 protein, decreased Beclin1 protein, decreased the ratio of LC3B-II/LC3B-I, and decreased the numbers of autophagosomes and autophagolysosomes in the SN tissues of mice. These effects were also abrogated by sCLU intervention. Activation of PI3K/AKT/mTOR signaling with MPTP inhibited autophagy in the SN of MPTP mice; however, sCLU treatment activated autophagy in MPTP-induced PD mice by inhibiting PI3K/AKT/mTOR signaling. These data indicated that sCLU treatment had a neuroprotective effect in an MPTP-induced model of PD.
Collapse
Affiliation(s)
- Dongxue Zhu
- Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Shenyang Zhang
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiaoying Wang
- Department of Ultrasound, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Chenghua Xiao
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Guiyun Cui
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xinxin Yang
- Department of Neurology, The Affifiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| |
Collapse
|
10
|
Li H, Zeng F, Huang C, Pu Q, Thomas ER, Chen Y, Li X. The potential role of glucose metabolism, lipid metabolism, and amino acid metabolism in the treatment of Parkinson's disease. CNS Neurosci Ther 2024; 30:e14411. [PMID: 37577934 PMCID: PMC10848100 DOI: 10.1111/cns.14411] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/15/2023] Open
Abstract
PURPOSE OF REVIEW Parkinson's disease (PD) is a common neurodegenerative disease, which can cause progressive deterioration of motor function causing muscle stiffness, tremor, and bradykinesia. In this review, we hope to describe approaches that can improve the life of PD patients through modifications of energy metabolism. RECENT FINDINGS The main pathological features of PD are the progressive loss of nigrostriatal dopaminergic neurons and the production of Lewy bodies. Abnormal aggregation of α-synuclein (α-Syn) leading to the formation of Lewy bodies is closely associated with neuronal dysfunction and degeneration. The main causes of PD are said to be mitochondrial damage, oxidative stress, inflammation, and abnormal protein aggregation. Presence of abnormal energy metabolism is another cause of PD. Many studies have found significant differences between neurodegenerative diseases and metabolic decompensation, which has become a biological hallmark of neurodegenerative diseases. SUMMARY In this review, we highlight the relationship between abnormal energy metabolism (Glucose metabolism, lipid metabolism, and amino acid metabolism) and PD. Improvement of key molecules in glucose metabolism, fat metabolism, and amino acid metabolism (e.g., glucose-6-phosphate dehydrogenase, triglycerides, and levodopa) might be potentially beneficial in PD. Some of these metabolic indicators may serve well during the diagnosis of PD. In addition, modulation of these metabolic pathways may be a potential target for the treatment and prevention of PD.
Collapse
Affiliation(s)
- Hangzhen Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | - Fancai Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | - Cancan Huang
- Department of DermatologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Qiqi Pu
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | | | - Yan Chen
- Department of DermatologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| |
Collapse
|
11
|
Morales-Carrizales DA, Gopar-Cuevas Y, Loera-Arias MDJ, Saucedo-Cardenas O, Montes de Oca-Luna R, Garcia-Garcia A, Rodriguez-Rocha H. A neuroprotective dose of trehalose is harmless to metabolic organs: comprehensive histopathological analysis of liver, pancreas, and kidney. Daru 2023; 31:135-144. [PMID: 37393413 PMCID: PMC10624785 DOI: 10.1007/s40199-023-00468-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/18/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND Trehalose is a non-reducing disaccharide synthesized by lower organisms. It has recently received special attention because of its neuroprotective properties by stimulating autophagy in Parkinson's disease (PD) models. Therefore, evaluating whether trehalose affects metabolic organs is vital to determine its neurotherapeutic safety. METHODS We validated the trehalose neuroprotective dosage in a PD model induced with intraperitoneal paraquat administration twice weekly for 7 weeks. One week before paraquat administration, mice were treated with trehalose in the drinking water and continued along with paraquat treatment. Histological and morphometrical analyses were conducted on the organs involved in trehalose metabolism, including the liver, pancreas, and kidney. RESULTS Paraquat-induced dopaminergic neuronal loss was significantly decreased by trehalose. After trehalose treatment, the liver morphology, the mononucleated/binucleated hepatocytes percentage, and sinusoidal diameter remained unchanged in each liver lobes. Endocrine and exocrine pancreas's histology was not affected, nor was any fibrotic process observed. The islet of Langerhans's structure was preserved when analyzing the area, the largest and smallest diameter, and circularity. Renal morphology remained undamaged, and no changes were identified within the glomerular basement membrane. The renal corpuscle structure did not suffer alterations in the Bowman's space, area, diameter, circularity, perimeter, and cellularity. Besides, the renal tubular structures's luminal area and internal and external diameter were preserved. CONCLUSION Our study demonstrates that systemic trehalose administration preserved the typical histological architecture of the organs involved in its metabolism, supporting its safety as a potential neuroprotective agent.
Collapse
Affiliation(s)
- Diego Armando Morales-Carrizales
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Yareth Gopar-Cuevas
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Maria de Jesus Loera-Arias
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Odila Saucedo-Cardenas
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Roberto Montes de Oca-Luna
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Aracely Garcia-Garcia
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico.
| | - Humberto Rodriguez-Rocha
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico.
| |
Collapse
|
12
|
Nápoles-Medina AY, Aguilar-Uscanga BR, Solís-Pacheco JR, Tejeda-Martínez AR, Ramírez-Jirano LJ, Urmeneta-Ortiz MF, Chaparro-Huerta V, Flores-Soto ME. Oral Administration of Lactobacillus Inhibits the Permeability of Blood-Brain and Gut Barriers in a Parkinsonism Model. Behav Neurol 2023; 2023:6686037. [PMID: 38025189 PMCID: PMC10653970 DOI: 10.1155/2023/6686037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
It has recently been shown that the administration of probiotics can modulate the microbiota-gut-brain axis and may have favorable effects in models of Parkinson's disease. In this study, we used a hemiparkinsonism model induced by the neurotoxin 6-OHDA to evaluate the efficacy of the administration of a four-week administration of a mixture containing the microorganisms Lactobacillus fermentum LH01, Lactobacillus reuteri LH03, and Lactobacillus plantarum LH05. The hemiparkinsonism model induced an increase in rotations in the apomorphine test, along with a decrease in the latency time to fall in the rotarod test on days 14 and 21 after surgery, respectively. The administration of probiotics was sufficient to improve this condition. The model also showed a decrease in tyrosine hydroxylase immunoreactivity in the striatum and the number of labeled cells in the substantia nigra, both of which were counteracted by the administration of probiotics. The permeability of the blood-brain barrier was increased in the model, but this effect was reversed by the probiotics for both brain regions. The gut barrier was permeated with the model, and this effect was reversed and dropped to lower levels than the control group after the administration of probiotics. Finally, lipid peroxidation showed a pattern of differences similar to that of permeabilities. The inhibition of the permeability of the blood-brain and gut barriers mediated by the administration of probiotics will likely provide protection by downregulating oxidative stress, thus affecting the rotarod test performance.
Collapse
Affiliation(s)
- Angélica Y. Nápoles-Medina
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada #800, Independencia Oriente, C.P. 44340 Guadalajara, Jalisco, Mexico
- Departamento de Farmacobiología, Laboratorio de Microbiología Industrial, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Blvd. Marcelino García Barragán # 1421, Olímpica, C.P. 44430 Guadalajara, Jalisco, Mexico
| | - Blanca R. Aguilar-Uscanga
- Departamento de Farmacobiología, Laboratorio de Microbiología Industrial, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Blvd. Marcelino García Barragán # 1421, Olímpica, C.P. 44430 Guadalajara, Jalisco, Mexico
| | - Josué R. Solís-Pacheco
- Departamento de Farmacobiología, Laboratorio de Microbiología Industrial, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Blvd. Marcelino García Barragán # 1421, Olímpica, C.P. 44430 Guadalajara, Jalisco, Mexico
| | - Aldo R. Tejeda-Martínez
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada #800, Independencia Oriente, C.P. 44340 Guadalajara, Jalisco, Mexico
| | - Luis J. Ramírez-Jirano
- División de Neurociencias, Centro de Investigación Biomédica Occidente (IMSS), Guadalajara, Mexico
| | - María F. Urmeneta-Ortiz
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada #800, Independencia Oriente, C.P. 44340 Guadalajara, Jalisco, Mexico
| | - Veronica Chaparro-Huerta
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada #800, Independencia Oriente, C.P. 44340 Guadalajara, Jalisco, Mexico
| | - Mario E. Flores-Soto
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Sierra Mojada #800, Independencia Oriente, C.P. 44340 Guadalajara, Jalisco, Mexico
| |
Collapse
|
13
|
Yap KH, Azmin S, Makpol S, Damanhuri HA, Mustapha M, Hamzah JC, Ibrahim NM. Profiling neuroprotective potential of trehalose in animal models of neurodegenerative diseases: a systematic review. Neural Regen Res 2023; 18:1179-1185. [PMID: 36453391 PMCID: PMC9838167 DOI: 10.4103/1673-5374.360164] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/24/2022] [Accepted: 10/13/2022] [Indexed: 11/27/2022] Open
Abstract
Trehalose, a unique nonreducing crystalline disaccharide, is a potential disease-modifying treatment for neurodegenerative diseases associated with protein misfolding and aggregation due to aging, intrinsic mutations, or autophagy dysregulation. This systematic review summarizes the effects of trehalose on its underlying mechanisms in animal models of selected neurodegenerative disorders (tau pathology, synucleinopathy, polyglutamine tract, and motor neuron diseases). All animal studies on neurodegenerative diseases treated with trehalose published in Medline (accessed via EBSCOhost) and Scopus were considered. Of the 2259 studies screened, 29 met the eligibility criteria. According to the SYstematic Review Center for Laboratory Animal Experiment (SYRCLE) risk of bias tool, we reported 22 out of 29 studies with a high risk of bias. The present findings support the purported role of trehalose in autophagic flux and protein refolding. This review identified several other lesser-known pathways, including modifying amyloid precursor protein processing, inhibition of reactive gliosis, the integrity of the blood-brain barrier, activation of growth factors, upregulation of the downstream antioxidant signaling pathway, and protection against mitochondrial defects. The absence of adverse events and improvements in the outcome parameters were observed in some studies, which supports the transition to human clinical trials. It is possible to conclude that trehalose exerts its neuroprotective effects through both direct and indirect pathways. However, heterogeneous methodologies and outcome measures across the studies rendered it impossible to derive a definitive conclusion. Translational studies on trehalose would need to clarify three important questions: 1) bioavailability with oral administration, 2) optimal time window to confer neuroprotective benefits, and 3) optimal dosage to confer neuroprotection.
Collapse
Affiliation(s)
- Kah Hui Yap
- Department of Medicine, UKM Medical Centre, Kuala Lumpur, Malaysia
| | - Shahrul Azmin
- Department of Medicine, UKM Medical Centre, Kuala Lumpur, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, UKM Medical Centre, Kuala Lumpur, Malaysia
| | | | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | | | | |
Collapse
|
14
|
Wang Y, Chen W, Han Y, Xu X, Yang A, Wei J, Hong D, Fang X, Chen T. Neuroprotective effect of engineered Clostridiumbutyricum-pMTL007-GLP-1 on Parkinson's disease mice models via promoting mitophagy. Bioeng Transl Med 2023; 8:e10505. [PMID: 37206220 PMCID: PMC10189449 DOI: 10.1002/btm2.10505] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/01/2023] [Accepted: 03/04/2023] [Indexed: 03/19/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease with limited treatment and no cure, hence, broadening PD drug spectrum is of great significance. At present, engineered microorganisms are attracting increasing attention. In this study, we constructed an engineered strain of Clostridium butyricum-GLP-1, a C. butyricum (a probiotic) that consistently expresses glucagon-like peptide-1 (GLP-1, a peptide-based hormone with neurological advantage) in anticipation of its use in PD treatment. We further investigated the neuroprotective mechanism of C. butyricum-GLP-1 on PD mice models induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. The results indicated that C. butyricum-GLP-1 could improve motor dysfunction and ameliorate neuropathological changes by increasing TH expression and reducing the expression of α-syn. Moreover, we confirmed that C. butyricum-GLP-1 improved microbiome imbalance of PD mice by decreasing the relative abundance of Bifidobacterium at the genus level, improved gut integrity, and upregulated the levels of GPR41/43. Surprisingly, we found it could exert its neuroprotective effects via promoting PINK1/Parkin mediated mitophagy and attenuating oxidative stress. Together, our work showed that C. butyricum-GLP-1 improves PD by promoting mitophagy, which provides an alternative therapeutic modality for PD.
Collapse
Affiliation(s)
- Yun Wang
- Department of NeurologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxi ProvinceP. R. China330006
| | - Wen‐jie Chen
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| | - Yi‐yang Han
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| | - Xuan Xu
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| | - Ai‐xia Yang
- Department of NeurologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxi ProvinceP. R. China330006
| | - Jing Wei
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| | - Dao‐jun Hong
- Department of NeurologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxi ProvinceP. R. China330006
| | - Xin Fang
- Department of NeurologyThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxi ProvinceP. R. China330006
| | - Ting‐tao Chen
- Institute of Translational MedicineNanchang UniversityNanchangJiangxi ProvinceP. R. China330031
| |
Collapse
|
15
|
Galbiati M, Meroni M, Boido M, Cescon M, Rusmini P, Crippa V, Cristofani R, Piccolella M, Ferrari V, Tedesco B, Casarotto E, Chierichetti M, Cozzi M, Mina F, Cicardi ME, Pedretti S, Mitro N, Caretto A, Risè P, Sala A, Lieberman AP, Bonaldo P, Pennuto M, Vercelli A, Poletti A. Bicalutamide and Trehalose Ameliorate Spinal and Bulbar Muscular Atrophy Pathology in Mice. Neurotherapeutics 2023; 20:524-545. [PMID: 36717478 PMCID: PMC10121997 DOI: 10.1007/s13311-023-01343-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2023] [Indexed: 02/01/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is characterized by motor neuron (MN) degeneration that leads to slowly progressive muscle weakness. It is considered a neuromuscular disease since muscle has a primary role in disease onset and progression. SBMA is caused by a CAG triplet repeat expansion in the androgen receptor (AR) gene. The translated poly-glutamine (polyQ) tract confers a toxic gain of function to the mutant AR altering its folding, causing its aggregation into intracellular inclusions, and impairing the autophagic flux. In an in vitro SBMA neuronal model, we previously showed that the antiandrogen bicalutamide and trehalose, a natural disaccharide stimulating autophagy, block ARpolyQ activation, reduce its nuclear translocation and toxicity and facilitate the autophagic degradation of cytoplasmic AR aggregates. Here, in a knock-in SBMA mouse model (KI AR113Q), we show that bicalutamide and trehalose ameliorated SBMA pathology. Bicalutamide reversed the formation of the AR insoluble forms in KI AR113Q muscle, preventing autophagic flux blockage. We demonstrated that apoptosis is activated in KI AR113Q muscle, and that both compounds prevented its activation. We detected a decrease of mtDNA and an increase of OXPHOS enzymes, already at early symptomatic stages; these alterations were reverted by trehalose. Overall, bicalutamide and/or trehalose led to a partial recovery of muscle morphology and function, and improved SBMA mouse motor behavior, inducing an extension of their survival. Thus, bicalutamide and trehalose, by counteracting ARpolyQ toxicity in skeletal muscle, are valuable candidates for future clinical trials in SBMA patients.
Collapse
Affiliation(s)
- Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy.
| | - Marco Meroni
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Marina Boido
- Department of Neuroscience Rita Levi Montalcini, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, Padua, Italy
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Margherita Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Marta Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Marta Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Francesco Mina
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Maria Elena Cicardi
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Weinberg ALS Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Silvia Pedretti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Nico Mitro
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy
| | - Anna Caretto
- Department of Neuroscience Rita Levi Montalcini, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Patrizia Risè
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milan, Italy
| | - Angelo Sala
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milan, Italy
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Padua, Italy
| | - Maria Pennuto
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Alessandro Vercelli
- Department of Neuroscience Rita Levi Montalcini, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Dipartimento di Eccellenza 2018-2027, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
16
|
Forouzanfar F, Vakilzadeh MM, Mehri A, Pourbagher-Shahri AM, Ganjali S, Abbasifard M, Sahebkar A. Anti-arthritic and Antioxidant Effects of Trehalose in an Experimental Model of Arthritis. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2023; 17:145-151. [PMID: 37622696 DOI: 10.2174/2772270817666230825093141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND The purpose of the present study was to study the potential anti-arthritic and antioxidant effects of trehalose in an experimental model of complete Freund's adjuvant (CFA)-induced arthritis. METHODS Arthritis was induced via subcutaneous injection of CFA (0.1) into the right footpad of each rat. Trehalose (10 mg/kg per day) and indomethacin (5 mg/kg) as a reference drug were intraperitoneally injected into CFA-induced arthritic rats from days 0 to 21. Changes in paw volume, pain responses, arthritic score, and oxidative/antioxidative parameters were determined. RESULTS Trehalose administration could significantly decrease arthritis scores (p <0.01) and paw edema (p <0.001), and significantly increase the nociceptive threshold (p <0.05) in CFA-induced arthritic rats. Trehalose also significantly reduced the pro-oxidant-antioxidant balance values when compared to CFA treatment alone. In addition, no significant difference was found between the trehalose group and indomethacin as a positive control group. CONCLUSION The current study suggests that trehalose has a protective effect against arthritis, which may be mediated by antioxidative effects of this disaccharide.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Mehri
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Shiva Ganjali
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Australia
| | - Mitra Abbasifard
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Tedesco B, Ferrari V, Cozzi M, Chierichetti M, Casarotto E, Pramaggiore P, Mina F, Piccolella M, Cristofani R, Crippa V, Rusmini P, Galbiati M, Poletti A. The role of autophagy-lysosomal pathway in motor neuron diseases. Biochem Soc Trans 2022; 50:1489-1503. [PMID: 36111809 PMCID: PMC9704526 DOI: 10.1042/bst20220778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 10/22/2023]
Abstract
Motor neuron diseases (MNDs) include a broad group of diseases in which neurodegeneration mainly affects upper and/or lower motor neurons (MNs). Although the involvement of specific MNs, symptoms, age of onset, and progression differ in MNDs, the main pathogenic mechanism common to most MNDs is represented by proteostasis alteration and proteotoxicity. This pathomechanism may be directly related to mutations in genes encoding proteins involved in the protein quality control system, particularly the autophagy-lysosomal pathway (ALP). Alternatively, proteostasis alteration can be caused by aberrant proteins that tend to misfold and to aggregate, two related processes that, over time, cannot be properly handled by the ALP. Here, we summarize the main ALP features, focusing on different routes utilized to deliver substrates to the lysosome and how the various ALP pathways intersect with the intracellular trafficking of membranes and vesicles. Next, we provide an overview of the mutated genes that have been found associated with MNDs, how these gene products are involved in different steps of ALP and related processes. Finally, we discuss how autophagy can be considered a valid therapeutic target for MNDs treatment focusing on traditional autophagy modulators and on emerging approaches to overcome their limitations.
Collapse
Affiliation(s)
- Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Marta Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Marta Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Paola Pramaggiore
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Francesco Mina
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Margherita Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
18
|
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
|
19
|
Mavroeidi P, Arvanitaki F, Vetsi M, Becker S, Vlachakis D, Jensen PH, Stefanis L, Xilouri M. Autophagy mediates the clearance of oligodendroglial SNCA/alpha-synuclein and TPPP/p25A in multiple system atrophy models. Autophagy 2022; 18:2104-2133. [PMID: 35000546 PMCID: PMC9466620 DOI: 10.1080/15548627.2021.2016256] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Accumulation of the neuronal protein SNCA/alpha-synuclein and of the oligodendroglial phosphoprotein TPPP/p25A within the glial cytoplasmic inclusions (GCIs) represents the key histophathological hallmark of multiple system atrophy (MSA). Even though the levels/distribution of both oligodendroglial SNCA and TPPP/p25A proteins are critical for disease pathogenesis, the proteolytic mechanisms involved in their turnover in health and disease remain poorly understood. Herein, by pharmacological and molecular modulation of the autophagy-lysosome pathway (ALP) and the proteasome we demonstrate that the endogenous oligodendroglial SNCA and TPPP/p25A are degraded mainly by the ALP in murine primary oligodendrocytes and oligodendroglial cell lines under basal conditions. We also identify a KFERQ-like motif in the TPPP/p25A sequence that enables its effective degradation via chaperone-mediated autophagy (CMA) in an in vitro system of rat brain lysosomes. Furthermore, in a MSA-like setting established by addition of human recombinant SNCA pre-formed fibrils (PFFs) as seeds of pathological SNCA, we thoroughly characterize the contribution of CMA and macroautophagy in particular, in the removal of the exogenously added and the seeded oligodendroglial SNCA pathological assemblies. We also show that PFF treatment impairs autophagic flux and that TPPP/p25A exerts an inhibitory effect on macroautophagy, while at the same time CMA is upregulated to remove the pathological SNCA species formed within oligodendrocytes. Finally, augmentation of CMA or macroautophagy accelerates the removal of the engendered pathological SNCA conformations further suggesting that autophagy targeting may represent a successful approach for the clearance of pathological SNCA and/or TPPP/p25A in the context of MSA.Abbreviations: 3MA: 3-methyladenine; ACTB: actin, beta; ALP: autophagy-lysosome pathway; ATG5: autophagy related 5; AR7: atypical retinoid 7; CMA: chaperone-mediated autophagy; CMV: cytomegalovirus; CTSD: cathepsin D; DAPI: 4',6-diamidino-2-phenylindole; DMEM: Dulbecco's modified Eagle's medium; Epox: epoxomicin; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GCIs: glial cytoplasmic inclusions; GFP: green fluorescent protein; HMW: high molecular weight; h: hours; HSPA8/HSC70: heat shock protein 8; LAMP1: lysosomal-associated membrane protein 1; LAMP2A: lysosomal-associated membrane protein 2A; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; mcherry: monomeric cherry; MFI: mean fluorescence intensity; mRFP: monomeric red fluorescent protein; MSA: multiple system atrophy; OLN: oligodendrocytes; OPCs: oligodendroglial progenitor cells; PBS: phosphate-buffered saline; PC12: pheochromocytoma cell line; PD: Parkinson disease; PFFs: pre-formed fibrils; PIs: protease inhibitors; PSMB5: proteasome (prosome, macropain) subunit, beta type 5; Rap: rapamycin; RFP: red fluorescent protein; Scr: scrambled; SDS: sodium dodecyl sulfate; SE: standard error; siRNAs: small interfering RNAs; SNCA: synuclein, alpha; SQSTM1: sequestosome 1; TPPP: tubulin polymerization promoting protein; TUBA: tubulin, alpha; UPS: ubiquitin-proteasome system; WT: wild type.
Collapse
Affiliation(s)
- Panagiota Mavroeidi
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Fedra Arvanitaki
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria Vetsi
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Stefan Becker
- Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Dimitrios Vlachakis
- Genetics and Computational Biology Group, Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Poul Henning Jensen
- DANDRITE-Danish Research Institute of Translational Neuroscience & Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Leonidas Stefanis
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece,1st Department of Neurology, Medical School, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece,CONTACT Maria Xilouri Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (Brfaa), 4 Soranou Efesiou Street, Athens11527, Greece
| |
Collapse
|
20
|
Pupyshev AB, Klyushnik TP, Akopyan AA, Singh SK, Tikhonova MA. Disaccharide Trehalose in Experimental Therapies for Neurodegenerative Disorders: Molecular Targets and Translational Potential. Pharmacol Res 2022; 183:106373. [PMID: 35907433 DOI: 10.1016/j.phrs.2022.106373] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 10/16/2022]
Abstract
Induction of autophagy is a prospective approach to the treatment of neurodegeneration. In the recent decade, trehalose attracted special attention. It is an autophagy inducer with negligible adverse effects and is approved for use in humans according to FDA requirements. Trehalose has a therapeutic effect in various experimental models of diseases. This glucose disaccharide with a flexible α-1-1'-glycosidic bond has unique properties: induction of mTOR-independent autophagy (with kinase AMPK as the main target) and a chaperone-like effect on proteins imparting them natural spatial structure. Thus, it can reduce the accumulation of neurotoxic aberrant/misfolded proteins. Trehalose has an anti-inflammatory effect and inhibits detrimental oxidative stress partially owing to the enhancement of endogenous antioxidant defense represented by the Nrf2 protein. The disaccharide activates lysosome and autophagosome biogenesis pathways through the protein factors TFEB and FOXO1. Here we review various mechanisms of the neuroprotective action of trehalose and touch on the possibility of pleiotropic effects. Current knowledge about specific features of trehalose pharmacodynamics is discussed. The neuroprotective effects of trehalose in animal models of major neurodegenerative disorders such as Alzheimer's, Parkinson's, and Huntington's diseases are examined too. Attention is given to translational transition to clinical trials of this drug, especially oral and parenteral routes of administration. Besides, the possibility of enhancing the therapeutic benefit via a combination of mTOR-dependent and mTOR-independent autophagy inducers is analyzed. In general, trehalose appears to be a promising multitarget tool for the inhibition of experimental neurodegeneration and requires thorough investigation of its clinical capabilities.
Collapse
Affiliation(s)
- Alexander B Pupyshev
- Scientific Research Institute of Neurosciences and Medicine (SRINM); Timakova Str. 4, Novosibirsk 630117, Russia.
| | - Tatyana P Klyushnik
- Mental Health Research Center, Kashirskoye shosse 34, Moscow 115522, Russia.
| | - Anna A Akopyan
- Scientific Research Institute of Neurosciences and Medicine (SRINM); Timakova Str. 4, Novosibirsk 630117, Russia.
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, Krishna Bhawan, 594 Kha/123, Shahinoor Colony, Nilmatha, Uttar Pradesh, Lucknow 226002, India.
| | - Maria A Tikhonova
- Scientific Research Institute of Neurosciences and Medicine (SRINM); Timakova Str. 4, Novosibirsk 630117, Russia.
| |
Collapse
|
21
|
Lang M, Pramstaller PP, Pichler I. Crosstalk of organelles in Parkinson's disease - MiT family transcription factors as central players in signaling pathways connecting mitochondria and lysosomes. Mol Neurodegener 2022; 17:50. [PMID: 35842725 PMCID: PMC9288732 DOI: 10.1186/s13024-022-00555-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/01/2022] [Indexed: 11/10/2022] Open
Abstract
Living organisms constantly need to adapt to their surrounding environment and have evolved sophisticated mechanisms to deal with stress. Mitochondria and lysosomes are central organelles in the response to energy and nutrient availability within a cell and act through interconnected mechanisms. However, when such processes become overwhelmed, it can lead to pathologies. Parkinson's disease (PD) is a common neurodegenerative disorder (NDD) characterized by proteinaceous intracellular inclusions and progressive loss of dopaminergic neurons, which causes motor and non-motor symptoms. Genetic and environmental factors may contribute to the disease etiology. Mitochondrial dysfunction has long been recognized as a hallmark of PD pathogenesis, and several aspects of mitochondrial biology are impaired in PD patients and models. In addition, defects of the autophagy-lysosomal pathway have extensively been observed in cell and animal models as well as PD patients' brains, where constitutive autophagy is indispensable for adaptation to stress and energy deficiency. Genetic and molecular studies have shown that the functions of mitochondria and lysosomal compartments are tightly linked and influence each other. Connections between these organelles are constituted among others by mitophagy, organellar dynamics and cellular signaling cascades, such as calcium (Ca2+) and mTOR (mammalian target of rapamycin) signaling and the activation of transcription factors. Members of the Microphthalmia-associated transcription factor family (MiT), including MITF, TFE3 and TFEB, play a central role in regulating cellular homeostasis in response to metabolic pressure and are considered master regulators of lysosomal biogenesis. As such, they are part of the interconnection between mitochondria and lysosome functions and therefore represent attractive targets for therapeutic approaches against NDD, including PD. The activation of MiT transcription factors through genetic and pharmacological approaches have shown encouraging results at ameliorating PD-related phenotypes in in vitro and in vivo models. In this review, we summarize the relationship between mitochondrial and autophagy-lysosomal functions in the context of PD etiology and focus on the role of the MiT pathway and its potential as pharmacological target against PD.
Collapse
Affiliation(s)
- Martin Lang
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy.
| | - Peter P Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy.,Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Irene Pichler
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| |
Collapse
|
22
|
Umeda-Miyara K, Miyara M, Sanoh S, Kotake Y. Trehalose decreases mRNA and protein expressions of c-Jun and JunB in human cervical cancer HeLa cells. J Biochem 2022; 172:177-187. [PMID: 35748379 DOI: 10.1093/jb/mvac051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 06/07/2022] [Indexed: 11/14/2022] Open
Abstract
Increasing evidence suggests that trehalose, a non-reducing disaccharide, ameliorates disease phenotypes by activating autophagy in animal models of various human diseases, including neurodegenerative diseases. Multiple in vitro studies suggest that activation of transcription factor EB, a master regulator of lysosomal biogenesis and autophagy genes, is a major contributor to trehalose-induced autophagy at later stages of exposure. However, underlying causes of trehalose-induced autophagy possibly occur at the early stage of the exposure period. In this study, we investigated the effects of short-term exposure of HeLa cells to trehalose on several signal transduction pathways to elucidate the initial events involved in its beneficial effects. Phospho-protein array analysis revealed that trehalose decreases levels of phosphorylated c-Jun, a component of the transcription factor activator protein-1, after 6 h. Trehalose also rapidly reduced mRNA expression levels of c-Jun and JunB, a member of the Jun family, within 1 h, resulting in a subsequent decrease in their protein levels. Future studies, exploring the interplay between decreased c-Jun and JunB protein levels and beneficial effects of trehalose may provide novel insights into the mechanisms of trehalose action.
Collapse
Affiliation(s)
- Kanae Umeda-Miyara
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| | - Masatsugu Miyara
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan.,Research Fellow of Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo 102-0083, Japan.,Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Gifu 501-1196, Japan
| | - Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| | - Yaichiro Kotake
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| |
Collapse
|
23
|
Wada SI, Arimura H, Nagayoshi M, Sawa R, Kubota Y, Matoba K, Hayashi C, Shibuya Y, Hatano M, Takehana Y, Ohba SI, Kobayashi Y, Watanabe T, Shibasaki M, Igarashi M. Rediscovery of 4-Trehalosamine as a Biologically Stable, Mass-Producible, and Chemically Modifiable Trehalose Analog. Adv Biol (Weinh) 2022; 6:e2101309. [PMID: 35297567 DOI: 10.1002/adbi.202101309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/02/2022] [Indexed: 01/27/2023]
Abstract
Nonreducing disaccharide trehalose is used as a stabilizer and humectant in various products and is a potential medicinal drug, showing curative effects on the animal models of various diseases. However, its use is limited as it is hydrolyzed by trehalase, a widely expressed enzyme in multiple organisms. Several trehalose analogs are prepared, including a microbial metabolite 4-trehalosamine, and their high biological stability is confirmed. For further analysis, 4-trehalosamine is selected as it shows high producibility. Compared with trehalose, 4-trehalosamine exhibits better or comparable protective activities and a high buffer capacity around the neutral pH. Another advantage of 4-trehalosamine is its chemical modifiability: simple reactions produce its various derivatives. Labeled probes and detergents are synthesized in one-pot reactions to exemplify the feasibility of their production, and their utility is confirmed for their respective applications. The labeled probes are used for mycobacterial staining. Although the derivative detergents can be effectively used in membrane protein research, long-chain detergents show 1000-3000-fold stronger autophagy-inducing activity in cultured cells than trehalose and are expected to become a drug lead and research reagent. These results indicate that 4-trehalosamine is a useful trehalose substitute for various purposes and a material to produce new useful derivative substances.
Collapse
Affiliation(s)
- Shun-Ichi Wada
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Honami Arimura
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Miho Nagayoshi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Ryuichi Sawa
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Yumiko Kubota
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Kazuaki Matoba
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Chigusa Hayashi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Yuko Shibuya
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Masaki Hatano
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Yasuhiro Takehana
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Shun-Ichi Ohba
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Yoshihiko Kobayashi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Takumi Watanabe
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Masakatsu Shibasaki
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Masayuki Igarashi
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| |
Collapse
|
24
|
Moon SH, Kwon Y, Huh YE, Choi HJ. Trehalose ameliorates prodromal non-motor deficits and aberrant protein accumulation in a rotenone-induced mouse model of Parkinson's disease. Arch Pharm Res 2022; 45:417-432. [PMID: 35618982 DOI: 10.1007/s12272-022-01386-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/18/2022] [Indexed: 02/03/2023]
Abstract
Trehalose has been recently revealed as an attractive candidate to prevent and modify Parkinson's disease (PD) progression by regulating autophagy; however, studies have only focused on the reduction of motor symptoms rather than the modulation of disease course from prodromal stage. This study aimed to evaluate whether trehalose has a disease-modifying effect at the prodromal stage before the onset of a motor deficit in 8-week-old male C57BL/6 mice exposed to rotenone. We found significant decrease in tyrosine hydroxylase immunoreactivity in the substantia nigra and motor dysfunction after 2 weeks rotenone treatment. Mice exposed to rotenone for a week showed an accumulation of protein aggregates in the brain and prodromal non-motor deficits, such as depression and olfactory dysfunction, prior to motor deficits. Trehalose significantly improved olfactory dysfunction and depressive-like behaviors and markedly reduced α-synuclein and p62 deposition in the brain. Trehalose further ameliorated motor impairment and loss of nigral tyrosine hydroxylase-positive cells in rotenone-treated mice. We demonstrated that prodromal non-motor signs in a rotenone-induced PD mouse model are associated with protein aggregate accumulation in the brain and that an autophagy inducer could be valuable to prevent PD progression from prodromal stage by regulating abnormal protein accumulation.
Collapse
Affiliation(s)
- Soung Hee Moon
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, Republic of Korea
| | - Yoonjung Kwon
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, Republic of Korea
| | - Young Eun Huh
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, 13488, Republic of Korea.
| | - Hyun Jin Choi
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, Republic of Korea.
| |
Collapse
|
25
|
Pupyshev AB, Belichenko VM, Tenditnik MV, Bashirzade AA, Dubrovina NI, Ovsyukova MV, Akopyan AA, Fedoseeva LA, Korolenko TA, Amstislavskaya TG, Tikhonova MA. Combined induction of mTOR-dependent and mTOR-independent pathways of autophagy activation as an experimental therapy for Alzheimer's disease-like pathology in a mouse model. Pharmacol Biochem Behav 2022; 217:173406. [DOI: 10.1016/j.pbb.2022.173406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/18/2022] [Accepted: 05/17/2022] [Indexed: 12/21/2022]
|
26
|
Ritzel RM, Li Y, Lei Z, Carter J, He J, Choi HMC, Khan N, Li H, Allen S, Lipinski MM, Faden AI, Wu J. Functional and transcriptional profiling of microglial activation during the chronic phase of TBI identifies an age-related driver of poor outcome in old mice. GeroScience 2022; 44:1407-1440. [PMID: 35451674 PMCID: PMC9213636 DOI: 10.1007/s11357-022-00562-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/01/2022] [Indexed: 12/14/2022] Open
Abstract
Elderly patients with traumatic brain injury (TBI) have greater mortality and poorer outcomes than younger individuals. The extent to which old age alters long-term recovery and chronic microglial activation after TBI is unknown, and evidence for therapeutic efficacy in aged mice is sorely lacking. The present study sought to identify potential inflammatory mechanisms underlying age-related outcomes late after TBI. Controlled cortical impact was used to induce moderate TBI in young and old male C57BL/6 mice. At 12 weeks post-injury, aged mice exhibited higher mortality, poorer functional outcomes, larger lesion volumes, and increased microglial activation. Transcriptomic analysis identified age- and TBI-specific gene changes consistent with a disease-associated microglial signature in the chronically injured brain, including those involved with complement, phagocytosis, and autophagy pathways. Dysregulation of phagocytic and autophagic function in microglia was accompanied by increased neuroinflammation in old mice. As proof-of-principle that these pathways have functional importance, we administered an autophagic enhancer, trehalose, in drinking water continuously for 8 weeks after TBI. Old mice treated with trehalose showed enhanced functional recovery and reduced microglial activation late after TBI compared to the sucrose control group. Our data indicate that microglia undergo chronic changes in autophagic regulation with both normal aging and TBI that are associated with poorer functional outcome. Enhancing autophagy may therefore be a promising clinical therapeutic strategy for TBI, especially in older patients.
Collapse
Affiliation(s)
- Rodney M. Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Yun Li
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Zhuofan Lei
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Jordan Carter
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Junyun He
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Harry M. C. Choi
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Niaz Khan
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Hui Li
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Samantha Allen
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Marta M. Lipinski
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Alan I. Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| |
Collapse
|
27
|
Grosso Jasutkar H, Oh SE, Mouradian MM. Therapeutics in the Pipeline Targeting α-Synuclein for Parkinson's Disease. Pharmacol Rev 2022; 74:207-237. [PMID: 35017177 PMCID: PMC11034868 DOI: 10.1124/pharmrev.120.000133] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and the fastest growing neurologic disease in the world, yet no disease-modifying therapy is available for this disabling condition. Multiple lines of evidence implicate the protein α-synuclein (α-Syn) in the pathogenesis of PD, and as such, there is intense interest in targeting α-Syn for potential disease modification. α-Syn is also a key pathogenic protein in other synucleionpathies, most commonly dementia with Lewy bodies. Thus, therapeutics targeting this protein will have utility in these disorders as well. Here we discuss the various approaches that are being investigated to prevent and mitigate α-Syn toxicity in PD, including clearing its pathologic aggregates from the brain using immunization strategies, inhibiting its misfolding and aggregation, reducing its expression level, enhancing cellular clearance mechanisms, preventing its cell-to-cell transmission within the brain and perhaps from the periphery, and targeting other proteins associated with or implicated in PD that contribute to α-Syn toxicity. We also discuss the therapeutics in the pipeline that harness these strategies. Finally, we discuss the challenges and opportunities for the field in the discovery and development of therapeutics for disease modification in PD. SIGNIFICANCE STATEMENT: PD is the second most common neurodegenerative disorder, for which disease-modifying therapies remain a major unmet need. A large body of evidence points to α-synuclein as a key pathogenic protein in this disease as well as in dementia with Lewy bodies, making it of leading therapeutic interest. This review discusses the various approaches being investigated and progress made to date toward discovering and developing therapeutics that would slow and stop progression of these disabling diseases.
Collapse
Affiliation(s)
- Hilary Grosso Jasutkar
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - Stephanie E Oh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - M Maral Mouradian
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| |
Collapse
|
28
|
Restoration of Parkinson's Disease-Like Deficits by Activating Autophagy through mTOR-Dependent and mTOR-Independent Mechanisms in Pharmacological and Transgenic Models of Parkinson's Disease in Mice. Bull Exp Biol Med 2021; 171:425-430. [PMID: 34542745 DOI: 10.1007/s10517-021-05242-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Indexed: 10/20/2022]
Abstract
We studied the possibilities of inhibition of neurodegeneration in MPTP-induced model of Parkinson's disease (PD) in C57Bl/6J mice and transgenic model of early PD stage (5-monthold B6.Cg-Tg(Prnp-SNCA*A53T)23Mkle/J mice) by autophagy activation through mTOR-dependent and mTOR-independent pathways with rapamycin and trehalose, respectively. Therapy with autophagy inducers in a "postponed" mode (7 days after MPTP intoxication) restored the expression of the dopaminergic neuron marker tyrosine hydroxylase and markedly improved cognitive function in the conditioned passive avoidance response (CPAR; fear memory). The transgenic model also showed an increase in the expression of tyrosine hydroxylase in the nigrostriatal system of the brain. An enhanced therapeutic effect of the combined treatment with the drugs was revealed on the expression of tyrosine hydroxylase, but not in the CPAR test. Thus, activation of both pathways of autophagy regulation in PD models with weakened neuroinflammation can restore the dopaminergic function of neurons and cognitive activity in mice.
Collapse
|
29
|
Long H, Qiu X, Cao L, Han R. Discovery of the signal pathways and major bioactive compounds responsible for the anti-hypoxia effect of Chinese cordyceps. JOURNAL OF ETHNOPHARMACOLOGY 2021; 277:114215. [PMID: 34033902 DOI: 10.1016/j.jep.2021.114215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/24/2021] [Accepted: 05/15/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hypoxia will cause an increase in the rate of fatigue and aging. Chinese cordyceps, a parasitic Thitarodes insect-Ophiocordyceps sinensis fungus complex in the Qinghai-Tibet Plateau, has long been used to ameliorate human conditions associated with aging and senescence, it is principally applied to treat fatigue, night sweating and other symptoms related to aging, and it may play the anti-aging and anti-fatigue effect by improving the body's hypoxia tolerance. AIMS OF THE STUDY The present study investigated the anti-hypoxia activity of Chinese cordyceps and explore the main corresponding signal pathways and bioactive compounds. MATERIALS AND METHODS In this study, network pharmacology analysis, molecular docking, cell and whole pharmacodynamic experiments were hired to study the major signal pathways and the bioactive compounds of Chinese cordyceps for anti-hypoxia activity. RESULTS 17 pathways which Chinese cordyceps acted on seemed to be related to the anti-hypoxia effect, and "VEGF signal pathway" was one of the most important pathway. Chinese cordyceps improved the survival rate and regulated the targets related VEGF signal pathway of H9C2 cells under hypoxia, and also had significant anti-hypoxia effects to mice. Chorioallantoic membrane model experiment showed that Chinese cordyceps and the main constituents of (9Z,12Z)-octadeca-9,12-dienoic acid and cerevisterol had significant angiogenic activity in hypoxia condition. CONCLUSION Based on the results of network pharmacology and molecular docking analysis, cell and whole pharmacodynamic experiments, promoting angiogenesis by regulating VEGF signal pathway might be one of the mechanisms of anti-hypoxia effect of Chinese cordyceps, (9Z, 12Z)-octadeca-9,12-dienoic acid and cerevisterol were considered as the major anti-hypoxia bioactive compounds in Chinese cordyceps.
Collapse
Affiliation(s)
- Hailin Long
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China.
| | - Xuehong Qiu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China.
| | - Li Cao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China.
| | - Richou Han
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China.
| |
Collapse
|
30
|
Trehalose Reduces Nerve Injury Induced Nociception in Mice but Negatively Affects Alertness. Nutrients 2021; 13:nu13092953. [PMID: 34578829 PMCID: PMC8469914 DOI: 10.3390/nu13092953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 07/31/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
Trehalose, a sugar from fungi, mimics starvation due to a block of glucose transport and induces Transcription Factor EB- mediated autophagy, likely supported by the upregulation of progranulin. The pro-autophagy effects help to remove pathological proteins and thereby prevent neurodegenerative diseases such as Alzheimer’s disease. Enhancing autophagy also contributes to the resolution of neuropathic pain in mice. Therefore, we here assessed the effects of continuous trehalose administration via drinking water using the mouse Spared Nerve Injury model of neuropathic pain. Trehalose had no effect on drinking, feeding, voluntary wheel running, motor coordination, locomotion, and open field, elevated plus maze, and Barnes Maze behavior, showing that it was well tolerated. However, trehalose reduced nerve injury-evoked nociceptive mechanical and thermal hypersensitivity as compared to vehicle. Trehalose had no effect on calcium currents in primary somatosensory neurons, pointing to central mechanisms of the antinociceptive effects. In IntelliCages, trehalose-treated mice showed reduced activity, in particular, a low frequency of nosepokes, which was associated with a reduced proportion of correct trials and flat learning curves in place preference learning tasks. Mice failed to switch corner preferences and stuck to spontaneously preferred corners. The behavior in IntelliCages is suggestive of sedative effects as a “side effect” of a continuous protracted trehalose treatment, leading to impairment of learning flexibility. Hence, trehalose diet supplements might reduce chronic pain but likely at the expense of alertness.
Collapse
|
31
|
Safren N, Tank EM, Malik AM, Chua JP, Santoro N, Barmada SJ. Development of a specific live-cell assay for native autophagic flux. J Biol Chem 2021; 297:101003. [PMID: 34303705 PMCID: PMC8368035 DOI: 10.1016/j.jbc.2021.101003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/27/2021] [Accepted: 07/21/2021] [Indexed: 01/09/2023] Open
Abstract
Autophagy is an evolutionarily conserved pathway mediating the breakdown of cellular proteins and organelles. Emphasizing its pivotal nature, autophagy dysfunction contributes to many diseases; nevertheless, development of effective autophagy modulating drugs is hampered by fundamental deficiencies in available methods for measuring autophagic activity or flux. To overcome these limitations, we introduced the photoconvertible protein Dendra2 into the MAP1LC3B locus of human cells via CRISPR/Cas9 genome editing, enabling accurate and sensitive assessments of autophagy in living cells by optical pulse labeling. We used this assay to perform high-throughput drug screens of four chemical libraries comprising over 30,000 diverse compounds, identifying several clinically relevant drugs and novel autophagy modulators. A select series of candidate compounds also modulated autophagy flux in human motor neurons modified by CRISPR/Cas9 to express GFP-labeled LC3. Using automated microscopy, we tested the therapeutic potential of autophagy induction in several distinct neuronal models of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). In doing so, we found that autophagy induction exhibited discordant effects, improving survival in disease models involving the RNA binding protein TDP-43, while exacerbating toxicity in neurons expressing mutant forms of UBQLN2 and C9ORF72 associated with familial ALS/FTD. These studies confirm the utility of the Dendra2-LC3 assay, while illustrating the contradictory effects of autophagy induction in different ALS/FTD subtypes.
Collapse
Affiliation(s)
- Nathaniel Safren
- Department of Neurology, University of Michigan, Ann Arbor Michigan, USA
| | - Elizabeth M Tank
- Department of Neurology, University of Michigan, Ann Arbor Michigan, USA
| | - Ahmed M Malik
- Department of Neurology, University of Michigan, Ann Arbor Michigan, USA
| | - Jason P Chua
- Department of Neurology, University of Michigan, Ann Arbor Michigan, USA
| | - Nicholas Santoro
- Center for Chemical Genomics, Life Sciences Institute, University of Michigan, Ann Arbor Michigan, USA
| | - Sami J Barmada
- Department of Neurology, University of Michigan, Ann Arbor Michigan, USA.
| |
Collapse
|
32
|
Kakoty V, K C S, Dubey SK, Yang CH, Taliyan R. Neuroprotective Effects of Trehalose and Sodium Butyrate on Preformed Fibrillar Form of α-Synuclein-Induced Rat Model of Parkinson's Disease. ACS Chem Neurosci 2021; 12:2643-2660. [PMID: 34197084 DOI: 10.1021/acschemneuro.1c00144] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Therapeutic options for Parkinson's disease (PD) are limited to a symptomatic approach, making it a global threat. Targeting aggregated alpha-synuclein (α-syn) clearance is a gold standard for ameliorating PD pathology, bringing autophagy into the limelight. Expression of autophagy related genes are under the regulation by histone modifications, however, its relevance in PD is yet to be established. Here, preformed fibrillar form (PFF) of α-syn was used to induce PD in wistar rats, which were thereafter subjected to treatment with trehalose (tre, 4g/kg, orally), a potent autophagy inducer and sodium butyrate (SB, 300 mg/kg, orally), a pan histone deacetylase inhibitor alone as well as in combination. The combination treatment significantly reduced motor deficits as evidenced after rotarod, narrow beam walk, and open field tests. Novel object location and recognition tests were performed to govern cognitive abnormality associated with advanced stage PD, which was overcome by the combination treatment. Additionally, with the combination, the level of pro-inflammatory cytokines were significantly reduced, along with elevated levels of dopamine and histone H3 acetylation. Further, mRNA analysis revealed that levels of certain autophagy related genes and proteins implicated in PD pathogenesis significantly improved after administration of both tre and SB. Immunofluorescence and H&E staining in the substantia nigra region mirrored a potential improvement after treatment with both tre and SB. Therefore, outcomes of the present study were adequate to prove that combinatorial efficacy with tre and SB may prove to be a formidable insight into ameliorating PD exacerbated by PFF α-syn as compared to its individual efficacy.
Collapse
Affiliation(s)
- Violina Kakoty
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, 333031 Pilani, Rajasthan, India
| | - Sarathlal K C
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, 333031 Pilani, Rajasthan, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, 333031 Pilani, Rajasthan, India
| | - Chih-Hao Yang
- Department of Pharmacology, Taipei Medical University, Taipei 110, Taiwan
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, 333031 Pilani, Rajasthan, India
| |
Collapse
|
33
|
Lee YS, Lai DM, Huang HJ, Lee-Chen GJ, Chang CH, Hsieh-Li HM, Lee GC. Prebiotic Lactulose Ameliorates the Cognitive Deficit in Alzheimer's Disease Mouse Model through Macroautophagy and Chaperone-Mediated Autophagy Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:2422-2437. [PMID: 33617267 DOI: 10.1021/acs.jafc.0c07327] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Lactulose, as a prebiotic, can be utilized by human gut microbiota and stimulate their growth. Although microbiota modulation has become an emerging approach to manage many diseases and can be achieved by the administration of prebiotics, fewer investigations have been carried out on the therapeutic mechanism of lactulose. Two trehalose analogs, lactulose and melibiose, were identified as having a neuroprotective effect in polyglutamine and Parkinson disease models. In this study, we examined lactulose and melibiose in a mouse primary hippocampal neuronal culture under the toxicity of oligomeric Aβ25-35. Lactulose was further tested in vivo because its effective concentration is lower than that of melibiose. Lactulose and trehalose were applied individually to mice before a bilateral intrahippocampal CA1 injection of oligomeric Aβ25-35. The administration of lactulose and trehalose attenuated the short-term memory and the learning retrieval of Alzheimer's disease (AD) mice. From a pathological analysis, we found that the pretreatment of lactulose and trehalose decreased neuroinflammation and increased the levels of the autophagic pathways. These results suggest that the neuroprotective effects of both lactulose and trehalose are achieved through anti-inflammation and autophagy. In addition, lactulose was better than trehalose in the enhancement of the synaptic protein expression level in AD mice. Therefore, lactulose could potentially be developed into a preventive and/or therapeutic disaccharide for AD.
Collapse
Affiliation(s)
- Yan-Suan Lee
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan
| | - Dar-Ming Lai
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Hei-Jen Huang
- Department of Nursing, Mackay Junior College of Medicine, Nursing and Management, Taipei 112, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan
| | - Ching-Hwa Chang
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan
| | - Guan-Chiun Lee
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan
| |
Collapse
|
34
|
Sinha P, Verma B, Ganesh S. Trehalose Ameliorates Seizure Susceptibility in Lafora Disease Mouse Models by Suppressing Neuroinflammation and Endoplasmic Reticulum Stress. Mol Neurobiol 2021; 58:1088-1101. [PMID: 33094475 DOI: 10.1007/s12035-020-02170-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 10/14/2020] [Indexed: 12/20/2022]
Abstract
Lafora disease (LD) is one of the progressive and fatal forms of a neurodegenerative disorder and is characterized by teenage-onset myoclonic seizures. Neuropathological changes in LD include the formation of abnormal glycogen as Lafora bodies, gliosis, and neuroinflammation. LD is caused by defects in the gene coding for phosphatase (laforin) or ubiquitin ligase (malin). Mouse models of LD, developed by targeted disruption of these two genes, develop most symptoms of LD and show increased susceptibility to induced seizures. Studies on mouse models also suggest that defective autophagy might contribute to LD etiology. In an attempt to understand the specific role of autophagy in LD pathogenesis, in this study, we fed LD animals with trehalose, an inducer of autophagy, for 3 months and looked at its effect on the neuropathology and seizure susceptibility. We demonstrate here that trehalose ameliorates gliosis, neuroinflammation, and endoplasmic reticulum stress and reduces susceptibility to induced seizures in LD animals. However, trehalose did not affect the formation of Lafora bodies, suggesting the epileptic phenotype in LD could be either secondary to or independent of Lafora bodies. Taken together, our results suggest that autophagy inducers can be considered as potential therapeutic molecules for Lafora disease.
Collapse
Affiliation(s)
- Priyanka Sinha
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India
| | - Bhupender Verma
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India
| | - Subramaniam Ganesh
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India.
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, India.
| |
Collapse
|
35
|
Cuevas E, Burks S, Raymick J, Robinson B, Gómez-Crisóstomo NP, Escudero-Lourdes C, Lopez AGG, Chigurupati S, Hanig J, Ferguson SA, Sarkar S. Tauroursodeoxycholic acid (TUDCA) is neuroprotective in a chronic mouse model of Parkinson's disease. Nutr Neurosci 2020; 25:1374-1391. [PMID: 33345721 DOI: 10.1080/1028415x.2020.1859729] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Parkinson's disease (PD) is a progressive motor disease of unknown etiology. Although neuroprotective ability of endogenous bile acid, tauroursodeoxycholic acid (TUDCA), shown in various diseases, including an acute model of PD,the potential therapeutic role of TUDCA in progressive models of PD that exhibit all aspects of PD has not been elucidated. In the present study, mice were assigned to one of four treatment groups: (1) Probenecid (PROB); (2) TUDCA, (3) MPTP + PROB (MPTPp); and (3) TUDCA + MPTPp. Methods: Markers for dopaminergic function, neuroinflammation, oxidative stress and autophagy were assessed using high performance liquid chromatography (HPLC), immunohistochemistry (IHC) and western blot (WB) methods. Locomotion was measured before and after treatments. Results: MPTPp decreased the expression of dopamine transporters (DAT) and tyrosine hydroxylase (TH), indicating dopaminergic damage, and induced microglial and astroglial activation as demonstrated by IHC analysis. MPTPp also decreased DA and its metabolites as demonstrated by HPLC analysis. Further, MPTPp-induced protein oxidation; increased LAMP-1 expression indicated autophagy and the promotion of alpha-synuclein (α-SYN) aggregation.. Discussion: Pretreatment with TUDCA protected against dopaminergic neuronal damage, prevented the microglial and astroglial activation, as well as the DA and DOPAC reductions caused by MPTPp. TUDCA by itself did not produce any significant change, with data similar to the negative control group. Pretreatment with TUDCA prevented protein oxidation and autophagy, in addition to inhibiting α-SYN aggregation. Although TUDCA pretreatment did not significantly affect locomotion, only acute treatment effects were measured, indicating more extensive assessments may be necessary to reveal potential therapeutic effects on behavior. Together, these results suggest that autophagy may be involved in the progression of PD and that TUDCA may attenuate these effects. The efficacy of TUDCA as a novel therapy in patients with PD clearly warrants further study.
Collapse
Affiliation(s)
- Elvis Cuevas
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | - Susan Burks
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | - James Raymick
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | - Bonnie Robinson
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | | | | | | | - Srinivasulu Chigurupati
- Office of Regulatory Affairs, Office of Regulatory Science, Food and Drug Administration, Rockville, MD, USA
| | - Joseph Hanig
- Office of Testing & Research, CDER/FDA, White Oak, MD, USA
| | - Sherry A Ferguson
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA
| |
Collapse
|
36
|
Atwood DJ, Brown CN, Holditch SJ, Pokhrel D, Thorburn A, Hopp K, Edelstein CL. The effect of trehalose on autophagy-related proteins and cyst growth in a hypomorphic Pkd1 mouse model of autosomal dominant polycystic kidney disease. Cell Signal 2020; 75:109760. [DOI: 10.1016/j.cellsig.2020.109760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/27/2022]
|
37
|
Fouka M, Mavroeidi P, Tsaka G, Xilouri M. In Search of Effective Treatments Targeting α-Synuclein Toxicity in Synucleinopathies: Pros and Cons. Front Cell Dev Biol 2020; 8:559791. [PMID: 33015057 PMCID: PMC7500083 DOI: 10.3389/fcell.2020.559791] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD), multiple system atrophy (MSA) and Dementia with Lewy bodies (DLB) represent pathologically similar, progressive neurodegenerative disorders characterized by the pathological aggregation of the neuronal protein α-synuclein. PD and DLB are characterized by the abnormal accumulation and aggregation of α-synuclein in proteinaceous inclusions within neurons named Lewy bodies (LBs) and Lewy neurites (LNs), whereas in MSA α-synuclein inclusions are mainly detected within oligodendrocytes named glial cytoplasmic inclusions (GCIs). The presence of pathologically aggregated α-synuclein along with components of the protein degradation machinery, such as ubiquitin and p62, in LBs and GCIs is considered to underlie the pathogenic cascade that eventually leads to the severe neurodegeneration and neuroinflammation that characterizes these diseases. Importantly, α-synuclein is proposed to undergo pathogenic misfolding and oligomerization into higher-order structures, revealing self-templating conformations, and to exert the ability of "prion-like" spreading between cells. Therefore, the manner in which the protein is produced, is modified within neural cells and is degraded, represents a major focus of current research efforts in the field. Given that α-synuclein protein load is critical to disease pathogenesis, the identification of means to limit intracellular protein burden and halt α-synuclein propagation represents an obvious therapeutic approach in synucleinopathies. However, up to date the development of effective therapeutic strategies to prevent degeneration in synucleinopathies is limited, due to the lack of knowledge regarding the precise mechanisms underlying the observed pathology. This review critically summarizes the recent developed strategies to counteract α-synuclein toxicity, including those aimed to increase protein degradation, to prevent protein aggregation and cell-to-cell propagation, or to engage antibodies against α-synuclein and discuss open questions and unknowns for future therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
38
|
Xiao M, Xiao ZJ, Yang B, Lan Z, Fang F. Blood-Brain Barrier: More Contributor to Disruption of Central Nervous System Homeostasis Than Victim in Neurological Disorders. Front Neurosci 2020; 14:764. [PMID: 32903669 PMCID: PMC7438939 DOI: 10.3389/fnins.2020.00764] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022] Open
Abstract
The blood-brain barrier (BBB) is a dynamic but solid shield in the cerebral microvascular system. It plays a pivotal role in maintaining central nervous system (CNS) homeostasis by regulating the exchange of materials between the circulation and the brain and protects the neural tissue from neurotoxic components as well as pathogens. Here, we discuss the development of the BBB in physiological conditions and then focus on the role of the BBB in cerebrovascular disease, including acute ischemic stroke and intracerebral hemorrhage, and neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Finally, we summarize recent advancements in the development of therapies targeting the BBB and outline future directions and outstanding questions in the field. We propose that BBB dysfunction not only results from, but is causal in the pathogenesis of neurological disorders; the BBB is more a contributor to the disruption of CNS homeostasis than a victim in neurological disorders.
Collapse
Affiliation(s)
- Minjia Xiao
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
- Department of Critical Care Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhi Jie Xiao
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Binbin Yang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ziwei Lan
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Fang Fang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Lin CH, Wei PC, Chen CM, Huang YT, Lin JL, Lo YS, Lin JL, Lin CY, Wu YR, Chang KH, Lee-Chen GJ. Lactulose and Melibiose Attenuate MPTP-Induced Parkinson's Disease in Mice by Inhibition of Oxidative Stress, Reduction of Neuroinflammation and Up-Regulation of Autophagy. Front Aging Neurosci 2020; 12:226. [PMID: 32848705 PMCID: PMC7396622 DOI: 10.3389/fnagi.2020.00226] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a common neurodegenerative disease characterized by the progressive loss of dopaminergic (DAergic) neurons in the ventral brain. A disaccharide trehalose has demonstrated the potential to mitigate the DAergic loss in disease models for PD. However, trehalose is rapidly hydrolyzed into glucose by trehalase in the intestine, limiting its potential for clinical practice. Here, we investigated the neuroprotective potential of two trehalase-indigestible analogs, lactulose and melibiose, in sub-chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. Treatment with MPTP generated significant motor deficits, inhibited dopamine levels, and down-regulated dopamine transporter (DAT) in the striatum. Expression levels of genes involved in anti-oxidative stress pathways, including superoxide dismutase 2 (SOD2), nuclear factor erythroid 2-related factor 2 (NRF2), and NAD(P)H dehydrogenase (NQO1) were also down-regulated. Meanwhile, expression of the oxidative stress marker 4-hydroxynonenal (4-HNE) was up-regulated along with increased microglia and astrocyte reactivity in the ventral midbrain following MPTP treatment. MPTP also reduced the activity of autophagy, evaluated by the autophagosomal marker microtubule-associated protein 1 light chain 3 (LC3)-II. Lactulose and melibiose significantly rescued motor deficits, increased dopamine in the striatum, reduced microglia and astrocyte reactivity as well as decreased levels of 4-HNE. Furthermore, lactulose and melibiose up-regulated SOD2, NRF2, and NQO1 levels, as well as enhanced the LC3-II/LC3-I ratio in the ventral midbrain with MPTP treatment. Our findings indicate the potential of lactulose and melibiose to protect DAergic neurons in PD.
Collapse
Affiliation(s)
- Chih-Hsin Lin
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Pei-Cih Wei
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | | | - Jia-Lan Lin
- Taipei First Girls High School, Taipei, Taiwan
| | - Yen-Shi Lo
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Jia-Li Lin
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
40
|
Kumar V, Lee JD, Coulson EJ, Woodruff TM. A validated quantitative method for the assessment of neuroprotective barrier impairment in neurodegenerative disease models. J Neurochem 2020; 158:807-817. [PMID: 32628780 DOI: 10.1111/jnc.15119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/08/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) are highly specialized structures that limit molecule entry from the blood and maintain homeostasis within the central nervous system (CNS). BBB and BSCB breakdown are associated with multiple neurodegenerative diseases. Given the key role of neuroprotective barrier impairment in neurodegeneration, it is important to identify an effective quantitative method to assess barrier integrity in animal models. In this study, we developed and validated a quantitative method for assessing BBB and BSCB integrity using sodium fluorescein, a compound that outperformed other fluorescent dyes. We demonstrated using this method that multiple CNS regions progressively increase in permeability in models of Huntington's disease and amyotrophic lateral sclerosis, whereas biphasic disruption occurred in a mouse model of Alzheimer's disease with disease progression. Collectively, we report a quantitative fluorometric marker with validated reproducible experimental methods that allows the effective assessment of BBB and BSCB integrity in animal models. This method could be useful to further the understanding of the contribution of these neuroprotective barriers to neurodegeneration processes.
Collapse
Affiliation(s)
- Vinod Kumar
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia
| | - John D Lee
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia
| | - Elizabeth J Coulson
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia.,Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Qld, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia.,Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Qld, Australia
| |
Collapse
|
41
|
Cristofani R, Crippa V, Cicardi ME, Tedesco B, Ferrari V, Chierichetti M, Casarotto E, Piccolella M, Messi E, Galbiati M, Rusmini P, Poletti A. A Crucial Role for the Protein Quality Control System in Motor Neuron Diseases. Front Aging Neurosci 2020; 12:191. [PMID: 32792938 PMCID: PMC7385251 DOI: 10.3389/fnagi.2020.00191] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
Motor neuron diseases (MNDs) are fatal diseases characterized by loss of motor neurons in the brain cortex, in the bulbar region, and/or in the anterior horns of the spinal cord. While generally sporadic, inherited forms linked to mutant genes encoding altered RNA/protein products have also been described. Several different mechanisms have been found altered or dysfunctional in MNDs, like the protein quality control (PQC) system. In this review, we will discuss how the PQC system is affected in two MNDs—spinal and bulbar muscular atrophy (SBMA) and amyotrophic lateral sclerosis (ALS)—and how this affects the clearance of aberrantly folded proteins, which accumulate in motor neurons, inducing dysfunctions and their death. In addition, we will discuss how the PQC system can be targeted to restore proper cell function, enhancing the survival of affected cells in MNDs.
Collapse
Affiliation(s)
- Riccardo Cristofani
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Valeria Crippa
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Maria Elena Cicardi
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy.,Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Barbara Tedesco
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Veronica Ferrari
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Marta Chierichetti
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Elena Casarotto
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Margherita Piccolella
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Elio Messi
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Mariarita Galbiati
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Paola Rusmini
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy
| | - Angelo Poletti
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Dipartimento di Eccellenza 2018-2022, Università degli Studi di Milano, Milan, Italy.,Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
42
|
Ji M, Niu S, Guo J, Mi H, Jiang P. Silencing RNF13 Alleviates Parkinson’s Disease – Like Problems in Mouse Models by Regulating the Endoplasmic Reticulum Stress–Mediated IRE1α-TRAF2-ASK1-JNK Pathway. J Mol Neurosci 2020; 70:1977-1986. [DOI: 10.1007/s12031-020-01599-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/15/2020] [Indexed: 12/11/2022]
|
43
|
Yon M, Decoville M, Sarou-Kanian V, Fayon F, Birman S. Spatially-resolved metabolic profiling of living Drosophila in neurodegenerative conditions using 1H magic angle spinning NMR. Sci Rep 2020; 10:9516. [PMID: 32528106 PMCID: PMC7289880 DOI: 10.1038/s41598-020-66218-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Drosophila flies are versatile animal models for the study of gene mutations in neuronal pathologies. Their small size allows performing in vivo Magic Angle Spinning (MAS) experiments to obtain high-resolution 1H nuclear magnetic resonance (NMR) spectra. Here, we use spatially-resolved 1H high-resolution MAS NMR to investigate in vivo metabolite contents in different segments of the fly body. A comparative study of metabolic changes was performed for three neurodegenerative disorders: two cell-specific neuronal and glial models of Huntington disease (HD) and a model of glutamate excitotoxicity. It is shown that these pathologies are characterized by specific and sometimes anatomically localized variations in metabolite concentrations. In two cases, the modifications of 1H MAS NMR spectra localized in fly heads were significant enough to allow the creation of a predictive model.
Collapse
Affiliation(s)
- Maxime Yon
- CEMHTI UPR3079, CNRS, Université d'Orléans, F-45071, Orléans, France
| | | | | | - Franck Fayon
- CEMHTI UPR3079, CNRS, Université d'Orléans, F-45071, Orléans, France
| | - Serge Birman
- GCRN-LPC UMR8249, CNRS, ESPCI Paris, PSL Research University, F-75005, Paris, France
| |
Collapse
|
44
|
Trehalose attenuates TGF-β1-induced fibrosis of hSCFs by activating autophagy. Mol Cell Biochem 2020; 470:175-188. [DOI: 10.1007/s11010-020-03760-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/16/2020] [Indexed: 01/29/2023]
|
45
|
Chen CM, Lin CH, Wu YR, Yen CY, Huang YT, Lin JL, Lin CY, Chen WL, Chao CY, Lee-Chen GJ, Su MT, Chang KH. Lactulose and Melibiose Inhibit α-Synuclein Aggregation and Up-Regulate Autophagy to Reduce Neuronal Vulnerability. Cells 2020; 9:cells9051230. [PMID: 32429337 PMCID: PMC7290909 DOI: 10.3390/cells9051230] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by selective dopaminergic (DAergic) neuronal degeneration in the substantia nigra (SN) and proteinaceous α-synuclein-positive Lewy bodies and Lewy neuritis. As a chemical chaperone to promote protein stability and an autophagy inducer to clear aggregate-prone proteins, a disaccharide trehalose has been reported to alleviate neurodegeneration in PD cells and mouse models. Its trehalase-indigestible analogs, lactulose and melibiose, also demonstrated potentials to reduce abnormal protein aggregation in spinocerebellar ataxia cell models. In this study, we showed the potential of lactulose and melibiose to inhibit α-synuclein aggregation using biochemical thioflavin T fluorescence, cryogenic transmission electron microscopy (cryo-TEM) and prokaryotic split Venus complementation assays. Lactulose and melibiose further reduced α-synuclein aggregation and associated oxidative stress, as well as protected cells against α-synuclein-induced neurotoxicity by up-regulating autophagy and nuclear factor, erythroid 2 like 2 (NRF2) pathway in DAergic neurons derived from SH-SY5Y cells over-expressing α-synuclein. Our findings strongly indicate the potential of lactulose and melibiose for mitigating PD neurodegeneration, offering new drug candidates for PD treatment.
Collapse
Affiliation(s)
- Chiung Mei Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (C.M.C.); (C.-H.L.); (Y.-R.W.); (W.-L.C.); (C.-Y.C.)
| | - Chih-Hsin Lin
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (C.M.C.); (C.-H.L.); (Y.-R.W.); (W.-L.C.); (C.-Y.C.)
| | - Yih-Ru Wu
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (C.M.C.); (C.-H.L.); (Y.-R.W.); (W.-L.C.); (C.-Y.C.)
| | - Chien-Yu Yen
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan;
| | - Yu-Ting Huang
- Taipei First Girls High School, Taipei 10045, Taiwan; (Y.-T.H.); (J.-L.L.)
| | - Jia-Lan Lin
- Taipei First Girls High School, Taipei 10045, Taiwan; (Y.-T.H.); (J.-L.L.)
| | - Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan;
| | - Wan-Ling Chen
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (C.M.C.); (C.-H.L.); (Y.-R.W.); (W.-L.C.); (C.-Y.C.)
| | - Chih-Ying Chao
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (C.M.C.); (C.-H.L.); (Y.-R.W.); (W.-L.C.); (C.-Y.C.)
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan;
- Correspondence: (G.-J.L.-C.); (M.-T.S.); (K.-H.C.); Tel.: +886-2-77346359 (G.-J.L.-C.); +886-2-77346244 (M.-T.S.); +886-3-3281200-8421 (K.-H.C.)
| | - Ming-Tsan Su
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan;
- Correspondence: (G.-J.L.-C.); (M.-T.S.); (K.-H.C.); Tel.: +886-2-77346359 (G.-J.L.-C.); +886-2-77346244 (M.-T.S.); +886-3-3281200-8421 (K.-H.C.)
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Taoyuan 33302, Taiwan; (C.M.C.); (C.-H.L.); (Y.-R.W.); (W.-L.C.); (C.-Y.C.)
- Correspondence: (G.-J.L.-C.); (M.-T.S.); (K.-H.C.); Tel.: +886-2-77346359 (G.-J.L.-C.); +886-2-77346244 (M.-T.S.); +886-3-3281200-8421 (K.-H.C.)
| |
Collapse
|
46
|
Santana MM, Paixão S, Cunha-Santos J, Silva TP, Trevino-Garcia A, Gaspar LS, Nóbrega C, Nobre RJ, Cavadas C, Greif H, Pereira de Almeida L. Trehalose alleviates the phenotype of Machado-Joseph disease mouse models. J Transl Med 2020; 18:161. [PMID: 32272938 PMCID: PMC7144062 DOI: 10.1186/s12967-020-02302-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/14/2020] [Indexed: 02/11/2023] Open
Abstract
Background Machado–Joseph disease (MJD), also known as spinocerebellar ataxia type 3, is the most common of the dominantly inherited ataxias worldwide and is characterized by mutant ataxin-3 aggregation and neuronal degeneration. There is no treatment available to block or delay disease progression. In this work we investigated whether trehalose, a natural occurring disaccharide widely used in food and cosmetic industry, would rescue biochemical, behavioral and neuropathological features of an in vitro and of a severe MJD transgenic mouse model. Methods Two MJD animal models, a lentiviral based and a transgenic model, were orally treated with 2% trehalose solution for a period of 4 and 30 weeks, respectively. Motor behavior (rotarod, grip strength and footprint patterns) was evaluated at different time points and neuropathological features were evaluated upon in-life phase termination. Results Trehalose-treated MJD mice equilibrated for a longer time in the rotarod apparatus and exhibited an improvement of ataxic gait in footprint analysis. Trehalose-mediated improvements in motor behaviour were associated with a reduction of the MJD-associated neuropathology, as MJD transgenic mice treated with trehalose presented preservation of cerebellar layers thickness and a decrease in the size of ataxin-3 aggregates in Purkinje cells. In agreement, an improvement of neuropathological features was also observed in the full length lentiviral-based mouse model of MJD submitted to 2% trehalose treatment. Conclusions The present study suggests trehalose as a safety pharmacological strategy to counteract MJD-associated behavioural and neuropathological impairments.
Collapse
Affiliation(s)
- Magda M Santana
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Susana Paixão
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal
| | - Janete Cunha-Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Teresa Pereira Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Allyson Trevino-Garcia
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Laetitia S Gaspar
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Clévio Nóbrega
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Department of Biomedical Sciences and Medicine, Centre for Biomedical Research (CBMR), Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| | - Rui Jorge Nobre
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | | | - Luís Pereira de Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal. .,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal. .,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
47
|
Borah P, Sanjeev A, Mattaparthi VSK. Computational investigation on the effect of Oleuropein aglycone on the α-synuclein aggregation. J Biomol Struct Dyn 2020; 39:1259-1270. [PMID: 32041489 DOI: 10.1080/07391102.2020.1728384] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is considered to be the second most common progressive neurodegenerative brain disorder after Alzheimer's disease, which is caused by misfolding and aggregation of Alpha-synuclein (α-synuclein). It is characterized by distinct aggregated fibrillary form of α-synuclein known as the Lewy bodies and Lewy neurites. The most promising approach to combat PD is to prevent the misfolding and subsequent aggregation of α-synuclein. Recently, Oleuropein aglycone (OleA) has been reported to stabilize the monomeric structure of α-synuclein, subsequently favoring the growth of nontoxic aggregates. Therefore, understanding the conformational dynamics of α-synuclein monomer in the presence of OleA is significant. Here, we have investigated the effect of OleA on the conformational dynamics and the aggregation propensity of α-synuclein using molecular dynamics simulation. From molecular dynamics trajectory analysis, we noticed that when OleA is bound to α-synuclein, the intramolecular distance between non-amyloid-β component domain and C-terminal domain of α-synuclein was increased, whereas long-range hydrophobic interactions between the two region were reduced. Oleuropein aglycone was found to interact with the N-terminal domain of α-synuclein, making this region unavailable for interaction with membranes and lipids for the formation of cellular toxic aggregates. From the binding-free energy analysis, we found binding affinity between α-synuclein and OleA to be indeed high (ΔGbind = -12.56 kcal mol-1 from MM-PBSA and ΔGbind = -27.41 kcal mol-1from MM-GBSA). Our findings in this study thus substantiate the effect of OleA on the structure and stabilization of α-synuclein monomer that subsequently favors the growth of stable and nontoxic aggregates.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Priyanka Borah
- Molecular Modelling and Simulation Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, India
| | - Airy Sanjeev
- Molecular Modelling and Simulation Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, India
| | - Venkata Satish Kumar Mattaparthi
- Molecular Modelling and Simulation Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, India
| |
Collapse
|
48
|
Suresh SN, Chakravorty A, Giridharan M, Garimella L, Manjithaya R. Pharmacological Tools to Modulate Autophagy in Neurodegenerative Diseases. J Mol Biol 2020; 432:2822-2842. [PMID: 32105729 DOI: 10.1016/j.jmb.2020.02.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/03/2020] [Accepted: 02/12/2020] [Indexed: 12/11/2022]
Abstract
Considerable evidences suggest a link between autophagy dysfunction, protein aggregation, and neurodegenerative diseases. Given that autophagy is a conserved intracellular housekeeping process, modulation of autophagy flux in various model organisms have highlighted its importance for maintaining proteostasis. In postmitotic cells such as neurons, compromised autophagy is sufficient to cause accumulation of ubiquitinated aggregates, neuronal dysfunction, degeneration, and loss of motor coordination-all hallmarks of neurodegenerative diseases. Reciprocally, enhanced autophagy flux augments cellular and organismal health, in addition to extending life span. These genetic studies not-withstanding a plethora of small molecule modulators of autophagy flux have been reported that alleviate disease symptoms in models of neurodegenerative diseases. This review summarizes the potential of such molecules to be, perhaps, one of the first autophagy drugs for treating these currently incurable diseases.
Collapse
Affiliation(s)
- S N Suresh
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Anushka Chakravorty
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India
| | - Mridhula Giridharan
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India
| | - Lakshmi Garimella
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India; Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India.
| |
Collapse
|
49
|
Barilar JO, Knezovic A, Perhoc AB, Homolak J, Riederer P, Salkovic-Petrisic M. Shared cerebral metabolic pathology in non-transgenic animal models of Alzheimer's and Parkinson's disease. J Neural Transm (Vienna) 2020; 127:231-250. [PMID: 32030485 PMCID: PMC7035309 DOI: 10.1007/s00702-020-02152-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/24/2020] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) and Alzheimer's disease (AD) are the most common chronic neurodegenerative disorders, characterized by motoric dysfunction or cognitive decline in the early stage, respectively, but often by both symptoms in the advanced stage. Among underlying molecular pathologies that PD and AD patients have in common, more attention is recently paid to the central metabolic dysfunction presented as insulin resistant brain state (IRBS) and altered cerebral glucose metabolism, both also explored in animal models of these diseases. This review aims to compare IRBS and alterations in cerebral glucose metabolism in representative non-transgenic animal PD and AD models. The comparison is based on the selectivity of the neurotoxins which cause experimental PD and AD, towards the cellular membrane and intracellular molecular targets as well as towards the selective neurons/non-neuronal cells, and the particular brain regions. Mitochondrial damage and co-expression of insulin receptors, glucose transporter-2 and dopamine transporter on the membrane of particular neurons as well as astrocytes seem to be the key points which are further discussed in a context of alterations in insulin signalling in the brain and its interaction with dopaminergic transmission, particularly regarding the time frame of the experimental AD/PD pathology appearance and the correlation with cognitive and motor symptoms. Such a perspective provides evidence on IRBS being a common underlying metabolic pathology and a contributor to neurodegenerative processes in representative non-transgenic animal PD and AD models, instead of being a direct cause of a particular neurodegenerative disorder.
Collapse
Affiliation(s)
- Jelena Osmanovic Barilar
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Ana Knezovic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Ana Babic Perhoc
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Peter Riederer
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Würzburg, Füchsleinstrasse 15, 97080, Würzburg, Germany
- Department and Research Unit of Psychiatry, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Melita Salkovic-Petrisic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia.
- Institute of Fundamental Clinical and Translational Neuroscience, Research Centre of Excellence, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, 10 000, Zagreb, Croatia.
| |
Collapse
|
50
|
Targeting Aggrephagy for the Treatment of Alzheimer's Disease. Cells 2020; 9:cells9020311. [PMID: 32012902 PMCID: PMC7072705 DOI: 10.3390/cells9020311] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/25/2020] [Accepted: 01/26/2020] [Indexed: 12/17/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common neurodegenerative diseases in older individuals with specific neuropsychiatric symptoms. It is a proteinopathy, pathologically characterized by the presence of misfolded protein (Aβ and Tau) aggregates in the brain, causing progressive dementia. Increasing studies have provided evidence that the defect in protein-degrading systems, especially the autophagy-lysosome pathway (ALP), plays an important role in the pathogenesis of AD. Recent studies have demonstrated that AD-associated protein aggregates can be selectively recognized by some receptors and then be degraded by ALP, a process termed aggrephagy. In this study, we reviewed the role of aggrephagy in AD development and discussed the strategy of promoting aggrephagy using small molecules for the treatment of AD.
Collapse
|