1
|
Mueller T, Jeffrey P, He Y, Ouyang X, Westbrook D, Darley-Usmar V, Goldberg MS, Volpicelli-Daley L, Zhang J. Alpha-synuclein preformed fibril-induced aggregation and dopaminergic cell death in cathepsin D overexpression and ZKSCAN3 knockout mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613763. [PMID: 39345452 PMCID: PMC11429970 DOI: 10.1101/2024.09.18.613763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
α-synuclein accumulation is recognized as a prominent feature in the majority of Parkinson's disease cases and also occurs in a broad range of neurodegenerative disorders including Alzheimer's disease. It has been shown that α-synuclein can spread from a donor cell to neighboring cells and thus propagate cellular damage, antagonizing the effectiveness of therapies such as transplantation of fetal or iPSC derived dopaminergic cells. As we and others previously have shown, insufficient lysosomal function due to genetic mutations or targeted disruption of cathepsin D can cause α-synuclein accumulation. We here investigated whether overexpression of cathepsin D or knockout (KO) of the transcriptional suppressor of lysosomal biogenesis ZKSCAN3 can attenuate propagation of α-synuclein aggregation and cell death. We examined dopaminergic neurodegeneration in the substantia nigra using stereology of tyrosine hydroxylase-immunoreactive cells 4 months and 6 months after intrastriatal injection of α-synuclein preformed fibrils or monomeric α-synuclein control in control, central nervous system (CNS)-cathepsin D overexpressing and CNS-specific ZKSCAN3 KO mice. We also examined pS129-α-synuclein aggregates in the substantia nigra, cortex, amygdala and striatum. The extent of dopaminergic neurodegeneration and pS129-α-synuclein aggregation in the brains of CNS-specific ZKSCAN3 knockout mice and CNS-cathepsin D overexpressing mice was similar to that observed in wild-type mice. Our results indicate that neither enhancing cathepsin D expression nor disrupting ZKSCAN3 in the CNS is sufficient to attenuate pS129-α-synuclein aggregate accumulation or dopaminergic neurodegeneration.
Collapse
|
2
|
Bigi A, Cascella R, Cecchi C. α-Synuclein oligomers and fibrils: partners in crime in synucleinopathies. Neural Regen Res 2023; 18:2332-2342. [PMID: 37282450 PMCID: PMC10360081 DOI: 10.4103/1673-5374.371345] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
The misfolding and aggregation of α-synuclein is the general hallmark of a group of devastating neurodegenerative pathologies referred to as synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. In such conditions, a range of different misfolded aggregates, including oligomers, protofibrils, and fibrils, are present both in neurons and glial cells. Growing experimental evidence supports the proposition that soluble oligomeric assemblies, formed during the early phases of the aggregation process, are the major culprits of neuronal toxicity; at the same time, fibrillar conformers appear to be the most efficient at propagating among interconnected neurons, thus contributing to the spreading of α-synuclein pathology. Moreover, α-synuclein fibrils have been recently reported to release soluble and highly toxic oligomeric species, responsible for an immediate dysfunction in the recipient neurons. In this review, we discuss the current knowledge about the plethora of mechanisms of cellular dysfunction caused by α-synuclein oligomers and fibrils, both contributing to neurodegeneration in synucleinopathies.
Collapse
Affiliation(s)
- Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| |
Collapse
|
3
|
Guo ZH, Khattak S, Rauf MA, Ansari MA, Alomary MN, Razak S, Yang CY, Wu DD, Ji XY. Role of Nanomedicine-Based Therapeutics in the Treatment of CNS Disorders. Molecules 2023; 28:1283. [PMID: 36770950 PMCID: PMC9921752 DOI: 10.3390/molecules28031283] [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: 10/19/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/31/2023] Open
Abstract
Central nervous system disorders, especially neurodegenerative diseases, are a public health priority and demand a strong scientific response. Various therapy procedures have been used in the past, but their therapeutic value has been insufficient. The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier is two of the barriers that protect the central nervous system (CNS), but are the main barriers to medicine delivery into the CNS for treating CNS disorders, such as brain tumors, Parkinson's disease, Alzheimer's disease, and Huntington's disease. Nanotechnology-based medicinal approaches deliver valuable cargos targeting molecular and cellular processes with greater safety, efficacy, and specificity than traditional approaches. CNS diseases include a wide range of brain ailments connected to short- and long-term disability. They affect millions of people worldwide and are anticipated to become more common in the coming years. Nanotechnology-based brain therapy could solve the BBB problem. This review analyzes nanomedicine's role in medication delivery; immunotherapy, chemotherapy, and gene therapy are combined with nanomedicines to treat CNS disorders. We also evaluated nanotechnology-based approaches for CNS disease amelioration, with the intention of stimulating the immune system by delivering medications across the BBB.
Collapse
Affiliation(s)
- Zi-Hua Guo
- Department of Neurology, Kaifeng Hospital of Traditional Chinese Medicine, No. 54 East Caizhengting St., Kaifeng 475000, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Mohd Ahmar Rauf
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research & Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Mohammad N. Alomary
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), P.O. Box 6086, Riyadh 11442, Saudi Arabia
| | - Sufyan Razak
- Dow Medical College, John Hopkins Medical Center, School of Medicine, Baltimore, MD 21205, USA
| | - Chang-Yong Yang
- School of Nursing and Health, Henan University, Kaifeng 475004, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- School of Stomatology, Henan University, Kaifeng 475004, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|
4
|
Paccione N, Rahmani M, Barcia E, Negro S. Antiparkinsonian Agents in Investigational Polymeric Micro- and Nano-Systems. Pharmaceutics 2022; 15:pharmaceutics15010013. [PMID: 36678642 PMCID: PMC9866990 DOI: 10.3390/pharmaceutics15010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Parkinson's disease (PD) is a devastating neurodegenerative disease characterized by progressive destruction of dopaminergic tissue in the central nervous system (CNS). To date, there is no cure for the disease, with current pharmacological treatments aimed at controlling the symptoms. Therefore, there is an unmet need for new treatments for PD. In addition to new therapeutic options, there exists the need for improved efficiency of the existing ones, as many agents have difficulties in crossing the blood-brain barrier (BBB) to achieve therapeutic levels in the CNS or exhibit inappropriate pharmacokinetic profiles, thereby limiting their clinical benefits. To overcome these limitations, an interesting approach is the use of drug delivery systems, such as polymeric microparticles (MPs) and nanoparticles (NPs) that allow for the controlled release of the active ingredients targeting to the desired site of action, increasing the bioavailability and efficacy of treatments, as well as reducing the number of administrations and adverse effects. Here we review the polymeric micro- and nano-systems under investigation as potential new therapies for PD.
Collapse
Affiliation(s)
- Nicola Paccione
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Mahdieh Rahmani
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-913941741
| | - Emilia Barcia
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Institute of Industrial Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Sofía Negro
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Institute of Industrial Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| |
Collapse
|
5
|
Schmidhuber S, Scheiblhofer S, Weiss R, Cserepes M, Tóvári J, Gadermaier G, Bezard E, De Giorgi F, Ichas F, Strunk D, Mandler M. A Novel C-Type Lectin Receptor-Targeted α-Synuclein-Based Parkinson Vaccine Induces Potent Immune Responses and Therapeutic Efficacy in Mice. Vaccines (Basel) 2022; 10:1432. [PMID: 36146508 PMCID: PMC9506002 DOI: 10.3390/vaccines10091432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
The progressive accumulation of misfolded α-synuclein (α-syn) in the brain is widely considered to be causal for the debilitating clinical manifestations of synucleinopathies including, most notably, Parkinson's disease (PD). Immunotherapies, both active and passive, against α-syn have been developed and are promising novel treatment strategies for such disorders. To increase the potency and specificity of PD vaccination, we created the 'Win the Skin Immune System Trick' (WISIT) vaccine platform designed to target skin-resident dendritic cells, inducing superior B and T cell responses. Of the six tested WISIT candidates, all elicited higher immune responses compared to conventional, aluminum adjuvanted peptide-carrier conjugate PD vaccines, in BALB/c mice. WISIT-induced antibodies displayed higher selectivity for α-syn aggregates than those induced by conventional vaccines. Additionally, antibodies induced by two selected candidates were shown to inhibit α-syn aggregation in a dose-dependent manner in vitro. To determine if α-syn fibril formation could also be inhibited in vivo, WISIT candidate type 1 (CW-type 1) was tested in an established synucleinopathy seeding model and demonstrated reduced propagation of synucleinopathy in vivo. Our studies provide proof-of-concept for the efficacy of the WISIT vaccine technology platform and support further preclinical and clinical development of this vaccine candidate.
Collapse
Affiliation(s)
- Sabine Schmidhuber
- Tridem Bioscience GmbH & CoKG, Campus Vienna Biocenter, Dr.-Bohrgasse 7, 1030 Vienna, Austria
| | - Sandra Scheiblhofer
- Tridem Bioscience GmbH & CoKG, Campus Vienna Biocenter, Dr.-Bohrgasse 7, 1030 Vienna, Austria
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Hellbrunner Straße 34, 5020 Salzburg, Austria
| | - Richard Weiss
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Hellbrunner Straße 34, 5020 Salzburg, Austria
| | - Mihály Cserepes
- KINETO Lab Ltd., Csillaghegyi út 19-21, H-1037 Budapest, Hungary
| | - József Tóvári
- KINETO Lab Ltd., Csillaghegyi út 19-21, H-1037 Budapest, Hungary
| | - Gabriele Gadermaier
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Hellbrunner Straße 34, 5020 Salzburg, Austria
| | - Erwan Bezard
- Motac Neuroscience, Alderley Park, Macclesfield SK10 4TF, UK
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Francesca De Giorgi
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Francois Ichas
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Dirk Strunk
- Cell Therapy Institute, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Markus Mandler
- Tridem Bioscience GmbH & CoKG, Campus Vienna Biocenter, Dr.-Bohrgasse 7, 1030 Vienna, Austria
| |
Collapse
|
6
|
Li Y, Gu Z, Lin S, Chen L, Dzreyan V, Eid M, Demyanenko S, He B. Histone Deacetylases as Epigenetic Targets for Treating Parkinson's Disease. Brain Sci 2022; 12:672. [PMID: 35625059 PMCID: PMC9140162 DOI: 10.3390/brainsci12050672] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is a chronic progressive neurodegenerative disease that is increasingly becoming a global threat to the health and life of the elderly worldwide. Although there are some drugs clinically available for treating PD, these treatments can only alleviate the symptoms of PD patients but cannot completely cure the disease. Therefore, exploring other potential mechanisms to develop more effective treatments that can modify the course of PD is still highly desirable. Over the last two decades, histone deacetylases, as an important group of epigenetic targets, have attracted much attention in drug discovery. This review focused on the current knowledge about histone deacetylases involved in PD pathophysiology and their inhibitors used in PD studies. Further perspectives related to small molecules that can inhibit or degrade histone deacetylases to treat PD were also discussed.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, China; (Y.L.); (Z.G.); (S.L.); (L.C.)
| | - Zhicheng Gu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, China; (Y.L.); (Z.G.); (S.L.); (L.C.)
| | - Shuxian Lin
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, China; (Y.L.); (Z.G.); (S.L.); (L.C.)
| | - Lei Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, China; (Y.L.); (Z.G.); (S.L.); (L.C.)
| | - Valentina Dzreyan
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Stachki Ave. 194/1, 344090 Rostov-on-Don, Russia; (V.D.); (M.E.)
| | - Moez Eid
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Stachki Ave. 194/1, 344090 Rostov-on-Don, Russia; (V.D.); (M.E.)
| | - Svetlana Demyanenko
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Stachki Ave. 194/1, 344090 Rostov-on-Don, Russia; (V.D.); (M.E.)
| | - Bin He
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, China; (Y.L.); (Z.G.); (S.L.); (L.C.)
| |
Collapse
|
7
|
Mechanisms of enhanced aggregation and fibril formation of Parkinson's disease-related variants of α-synuclein. Sci Rep 2022; 12:6770. [PMID: 35474118 PMCID: PMC9043213 DOI: 10.1038/s41598-022-10789-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/08/2022] [Indexed: 12/18/2022] Open
Abstract
Aggregation of α-synuclein (α-syn) into amyloid fibrils is closely associated with Parkinson’s disease (PD). Familial mutations or posttranslational truncations in α-syn are known as risk factor for PD. Here, we examined the effects of the PD-related A30P or A53T point mutation and C-terminal 123–140 or 104–140 truncation on the aggregating property of α-syn based on the kinetic and thermodynamic analyses. Thioflavin T fluorescence measurements indicated that A53T, Δ123‒140, and Δ104–140 variants aggregated faster than WT α-syn, in which the A53T mutation markedly increases nucleation rate whereas the Δ123‒140 or Δ104‒140 truncation significantly increases both nucleation and fibril elongation rates. Ultracentrifugation and western blotting analyses demonstrated that these mutations or truncations promote the conversion of monomer to aggregated forms of α-syn. Analysis of the dependence of aggregation reaction of α-syn variants on the monomer concentration suggested that the A53T mutation enhances conversion of monomers to amyloid nuclei whereas the C-terminal truncations, especially the Δ104–140, enhance autocatalytic aggregation on existing fibrils. In addition, thermodynamic analysis of the kinetics of nucleation and fibril elongation of α-syn variants indicated that both nucleation and fibril elongation of WT α-syn are enthalpically and entropically unfavorable. Interestingly, the unfavorable activation enthalpy of nucleation greatly decreases for the A53T and becomes reversed in sign for the C-terminally truncated variants. Taken together, our results indicate that the A53T mutation and the C-terminal truncation enhance α-syn aggregation by reducing unfavorable activation enthalpy of nucleation, and the C-terminal truncation further triggers the autocatalytic fibril elongation on the fibril surfaces.
Collapse
|
8
|
Onaolapo OJ, Odeniyi AO, Onaolapo AY. Parkinson's Disease: Is there a Role for Dietary and Herbal Supplements? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 20:343-365. [PMID: 33602107 DOI: 10.2174/1871527320666210218082954] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/19/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
Parkinson's Disease (PD) is characterised by degeneration of the neurons of the nigrostriatal dopaminergic pathway of the brain. The pharmacological cornerstone of PD management is mainly the use of dopamine precursors, dopamine receptor agonists, and agents that inhibit the biochemical degradation of dopamine. While these drugs initially provide relief to the symptoms and improve the quality of life of the patients, progression of the underlying pathological processes, such as oxidative stress and neuroinflammation (which have been strongly associated with PD and other neurodegenerative disorders), eventually reduce their benefits, making further benefits achievable, only at high doses due to which the magnitude and frequency of side-effects are amplified. Also, while it is becoming obvious that mainstream pharmacological agents may not always provide the much-needed answer, the question remains what succour can nature provide through dietary supplements, nutraceuticals and herbal remedies? This narrative review examines current literature for evidence of the possible roles (if any) of nutraceuticals, dietary supplements and herbal remedies in the prevention or management of PD by examining how these compounds could modulate key factors and pathways that are crucial to the pathogenesis and/or progression of PD. The likely limitations of this approach and its possible future roles in PD prevention and management are also considered.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Ademola O Odeniyi
- Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Adejoke Y Onaolapo
- Behavioural Neuroscience Unit, Neurobiology Subdivision, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| |
Collapse
|
9
|
Mahía A, Peña-Díaz S, Navarro S, José Galano-Frutos J, Pallarés I, Pujols J, Díaz-de-Villegas MD, Gálvez JA, Ventura S, Sancho J. Design, synthesis and structure-activity evaluation of novel 2-pyridone-based inhibitors of α-synuclein aggregation with potentially improved BBB permeability. Bioorg Chem 2021; 117:105472. [PMID: 34775206 DOI: 10.1016/j.bioorg.2021.105472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/20/2021] [Accepted: 11/03/2021] [Indexed: 01/07/2023]
Abstract
The treatment of Parkinson's disease (PD), the second most common neurodegenerative human disorder, continues to be symptomatic. Development of drugs able to stop or at least slowdown PD progression would benefit several million people worldwide. SynuClean-D is a low molecular weight 2-pyridone-based promising drug candidate that inhibits the aggregation of α-synuclein in human cultured cells and prevents degeneration of dopaminergic neurons in a Caenorhabditis elegans model of PD. Improving SynuClean-D pharmacokinetic/pharmacodynamic properties, performing structure/activity studies and testing its efficacy in mammalian models of PD requires the use of gr-amounts of the compound. However, not enough compound is on sale, and no synthetic route has been reported until now, which hampers the molecule progress towards clinical trials. To circumvent those problems, we describe here an efficient and economical route that enables the synthesis of SynuClean-D with good yields as well as the synthesis of SynuClean-D derivatives. Structure-activity comparison of the new compounds with SynuClean-D reveals the functional groups of the molecule that can be disposed of without activity loss and those that are crucial to interfere with α-synuclein aggregation. Several of the derivatives obtained retain the parent's compound excellent in vitro anti-aggregative activity, without compromising its low toxicity. Computational predictions and preliminary testing indicate that the blood brain barrier (BBB) permeability of SynuClean-D is low. Importantly, several of the newly designed and obtained active derivatives are predicted to display good BBB permeability. The synthetic route developed here will facilitate their synthesis for BBB permeability determination and for efficacy testing in mammalian models of PD.
Collapse
Affiliation(s)
- Alejandro Mahía
- Departamento de Química Orgánica, Facultad de Ciencias, University of Zaragoza, 50009 Zaragoza, Spain; Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain
| | - Samuel Peña-Díaz
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Susanna Navarro
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Juan José Galano-Frutos
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, 50009 Zaragoza, Spain
| | - Irantzu Pallarés
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Jordi Pujols
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - María D Díaz-de-Villegas
- Departamento de Química Orgánica, Facultad de Ciencias, University of Zaragoza, 50009 Zaragoza, Spain; Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC-University of Zaragoza, 50009 Zaragoza, Spain
| | - José A Gálvez
- Departamento de Química Orgánica, Facultad de Ciencias, University of Zaragoza, 50009 Zaragoza, Spain; Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC-University of Zaragoza, 50009 Zaragoza, Spain.
| | - Salvador Ventura
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; ICREA, 08010 Barcelona, Spain.
| | - Javier Sancho
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, 50009 Zaragoza, Spain; Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain.
| |
Collapse
|
10
|
Zhang K, Zhu S, Li J, Jiang T, Feng L, Pei J, Wang G, Ouyang L, Liu B. Targeting autophagy using small-molecule compounds to improve potential therapy of Parkinson's disease. Acta Pharm Sin B 2021; 11:3015-3034. [PMID: 34729301 PMCID: PMC8546670 DOI: 10.1016/j.apsb.2021.02.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/28/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD), known as one of the most universal neurodegenerative diseases, is a serious threat to the health of the elderly. The current treatment has been demonstrated to relieve symptoms, and the discovery of new small-molecule compounds has been regarded as a promising strategy. Of note, the homeostasis of the autolysosome pathway (ALP) is closely associated with PD, and impaired autophagy may cause the death of neurons and thereby accelerating the progress of PD. Thus, pharmacological targeting autophagy with small-molecule compounds has been drawn a rising attention so far. In this review, we focus on summarizing several autophagy-associated targets, such as AMPK, mTORC1, ULK1, IMPase, LRRK2, beclin-1, TFEB, GCase, ERRα, C-Abelson, and as well as their relevant small-molecule compounds in PD models, which will shed light on a clue on exploiting more potential targeted small-molecule drugs tracking PD treatment in the near future.
Collapse
Key Words
- 3-MA, 3-methyladenine
- 5-HT2A, Serotonin 2A
- 5-HT2C, serotonin 2C
- A2A, adenosine 2A
- AADC, aromatic amino acid decarboxylase
- ALP, autophagy-lysosomal pathway
- AMPK, 5ʹAMP-activated protein kinase
- ATG, autophagy related protein
- ATP13A2, ATPase cation transporting 13A2
- ATTEC, autophagosome-tethering compound
- AUC, the area under the curve
- AUTAC, autophagy targeting chimera
- Autophagy
- BAF, bafilomycinA1
- BBB, blood−brain barrier
- CL, clearance rate
- CMA, chaperone-mediated autophagy
- CNS, central nervous system
- COMT, catechol-O-methyltransferase
- DA, dopamine
- DAT, dopamine transporter
- DJ-1, Parkinson protein 7
- DR, dopamine receptor
- ER, endoplasmic reticulum
- ERRα, estrogen-related receptor alpha
- F, oral bioavailability
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- GBA, glucocerebrosidase β acid
- GWAS, genome-wide association study
- HDAC6, histone deacetylase 6
- HSC70, heat shock cognate 71 kDa protein
- HSPA8, heat shock 70 kDa protein 8
- IMPase, inositol monophosphatase
- IPPase, inositol polyphosphate 1-phosphatase
- KI, knockin
- LAMP2A, lysosome-associated membrane protein 2 A
- LC3, light chain 3
- LIMP-2, lysosomal integrated membrane protein-2
- LRRK2, leucine-rich repeat sequence kinase 2
- LRS, leucyl-tRNA synthetase
- LUHMES, lund human mesencephalic
- Lamp2a, type 2A lysosomal-associated membrane protein
- MAO-B, monoamine oxidase B
- MPP+, 1-methyl-4-phenylpyridinium
- MPTP, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine
- MYCBP2, MYC-binding protein 2
- NMDA, N-methyl-d-aspartic acid
- ONRs, orphan nuclear receptors
- PD therapy
- PD, Parkinson's disease
- PDE4, phosphodiesterase 4
- PI3K, phosphatidylinositol 3-kinase
- PI3P, phosphatidylinositol 3-phosphate
- PINK1, PTEN-induced kinase 1
- PLC, phospholipase C
- PREP, prolyl oligopeptidase
- Parkin, parkin RBR E3 ubiquitin−protein ligase
- Parkinson's disease (PD)
- ROS, reactive oxygen species
- SAR, structure–activity relationship
- SAS, solvent accessible surface
- SN, substantia nigra
- SNCA, α-synuclein gene
- SYT11, synaptotagmin 11
- Small-molecule compound
- TFEB, transcription factor EB
- TSC2, tuberous sclerosis complex 2
- Target
- ULK1, UNC-51-like kinase 1
- UPS, ubiquitin−proteasome system
- mAChR, muscarinic acetylcholine receptor
- mTOR, the mammalian target of rapamycin
- α-syn, α-synuclein
Collapse
|
11
|
Khan A, Johnson R, Wittmer C, Maile M, Tatsukawa K, Wong JL, Gill MB, Stocking EM, Natala SR, Paulino AD, Bowden-Verhoek JK, Wrasidlo W, Masliah E, Bonhaus DW, Price DL. NPT520-34 improves neuropathology and motor deficits in a transgenic mouse model of Parkinson's disease. Brain 2021; 144:3692-3709. [PMID: 34117864 DOI: 10.1093/brain/awab214] [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: 01/06/2021] [Revised: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/09/2022] Open
Abstract
NPT520-34 is a clinical-stage, small molecule being developed for the treatment of Parkinson's disease and other neurodegenerative disorders. The therapeutic potential of NPT520-34 was first suggested by findings from cell-based assays of alpha-synuclein (ASYN) clearance. As reported here, NPT520-34 was subsequently evaluated for therapeutically relevant actions in a transgenic animal model of Parkinson's disease that overexpresses human ASYN and in an acute lipopolysaccharide (LPS)-challenge model using wild-type mice. Daily administration of NPT520-34 to mThy1-ASYN (Line 61) transgenic mice for one or three months resulted in reduced ASYN pathology, reduced expression of markers of neuroinflammation, and improvements in multiple indices of motor function. In an LPS-challenge model using wild-type mice, a single-dose of NPT520-34 reduced LPS-evoked increases in the expression of several pro-inflammatory cytokines in plasma. These findings demonstrate the beneficial effects of NPT520-34 on both inflammation and protein-pathology endpoints, with consequent improvements in motor function in an animal model of Parkinson's disease. These findings further suggest that NPT520-34 may have two complementary actions: (1) to increase the clearance of neurotoxic protein aggregates and (2) to directly attenuate inflammation. NPT520-34 treatment may thereby address two of the predominate underlying pathophysiological aspects of neurodegenerative disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Asma Khan
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Robert Johnson
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Carrie Wittmer
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Michelle Maile
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Keith Tatsukawa
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Julian L Wong
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Martin B Gill
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Emily M Stocking
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Srinivasa R Natala
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Amy D Paulino
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Jon K Bowden-Verhoek
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Wolfgang Wrasidlo
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Eliezer Masliah
- Departments of Neuroscience and Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Douglas W Bonhaus
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Diana L Price
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| |
Collapse
|
12
|
Duwa R, Jeong JH, Yook S. Development of immunotherapy and nanoparticles-based strategies for the treatment of Parkinson’s disease. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021. [DOI: 10.1007/s40005-021-00521-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
13
|
Hardenberg MC, Sinnige T, Casford S, Dada ST, Poudel C, Robinson EA, Fuxreiter M, Kaminksi CF, Kaminski Schierle GS, Nollen EAA, Dobson CM, Vendruscolo M. Observation of an α-synuclein liquid droplet state and its maturation into Lewy body-like assemblies. J Mol Cell Biol 2021; 13:282-294. [PMID: 33386842 DOI: 10.1093/jmcb/mjaa075] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/09/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Misfolded α-synuclein is a major component of Lewy bodies, which are a hallmark of Parkinson's disease (PD). A large body of evidence shows that α-synuclein can aggregate into amyloid fibrils, but the relationship between α-synuclein self-assembly and Lewy body formation remains unclear. Here, we show, both in vitro and in a Caenorhabditis elegans model of PD, that α-synuclein undergoes liquid‒liquid phase separation by forming a liquid droplet state, which converts into an amyloid-rich hydrogel with Lewy-body-like properties. This maturation process towards the amyloid state is delayed in the presence of model synaptic vesicles in vitro. Taken together, these results suggest that the formation of Lewy bodies may be linked to the arrested maturation of α-synuclein condensates in the presence of lipids and other cellular components.
Collapse
Affiliation(s)
- Maarten C Hardenberg
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Tessa Sinnige
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Sam Casford
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Samuel T Dada
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Chetan Poudel
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Elizabeth A Robinson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Monika Fuxreiter
- MTA-DE Laboratory of Protein Dynamics, Department of Biochemistry and Molecular Biology, University of Debrecen, Hungary
| | - Clemens F Kaminksi
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | | | - Ellen A A Nollen
- European Research Institute for the Biology of Aging, University Medical Center Groningen, University of Groningen, Groningen 9713 AV, The Netherlands
| | - Christopher M Dobson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| |
Collapse
|
14
|
Staats R, Michaels TCT, Flagmeier P, Chia S, Horne RI, Habchi J, Linse S, Knowles TPJ, Dobson CM, Vendruscolo M. Screening of small molecules using the inhibition of oligomer formation in α-synuclein aggregation as a selection parameter. Commun Chem 2020; 3:191. [PMID: 36703335 PMCID: PMC9814678 DOI: 10.1038/s42004-020-00412-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/15/2020] [Indexed: 01/29/2023] Open
Abstract
The aggregation of α-synuclein is a central event in Parkinsons's disease and related synucleinopathies. Since pharmacologically targeting this process, however, has not yet resulted in approved disease-modifying treatments, there is an unmet need of developing novel methods of drug discovery. In this context, the use of chemical kinetics has recently enabled accurate quantifications of the microscopic steps leading to the proliferation of protein misfolded oligomers. As these species are highly neurotoxic, effective therapeutic strategies may be aimed at reducing their numbers. Here, we exploit this quantitative approach to develop a screening strategy that uses the reactive flux toward α-synuclein oligomers as a selection parameter. Using this approach, we evaluate the efficacy of a library of flavone derivatives, identifying apigenin as a compound that simultaneously delays and reduces the formation of α-synuclein oligomers. These results demonstrate a compound selection strategy based on the inhibition of the formation of α-synuclein oligomers, which may be key in identifying small molecules in drug discovery pipelines for diseases associated with α-synuclein aggregation.
Collapse
Affiliation(s)
- Roxine Staats
- grid.5335.00000000121885934Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| | - Thomas C. T. Michaels
- grid.5335.00000000121885934Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK ,grid.38142.3c000000041936754XPaulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 USA
| | - Patrick Flagmeier
- grid.5335.00000000121885934Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| | - Sean Chia
- grid.5335.00000000121885934Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| | - Robert I. Horne
- grid.5335.00000000121885934Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| | - Johnny Habchi
- grid.5335.00000000121885934Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| | - Sara Linse
- grid.4514.40000 0001 0930 2361Department of Chemistry, Division for Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden
| | - Tuomas P. J. Knowles
- grid.5335.00000000121885934Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| | - Christopher M. Dobson
- grid.5335.00000000121885934Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| | - Michele Vendruscolo
- grid.5335.00000000121885934Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW UK
| |
Collapse
|
15
|
Tira R, De Cecco E, Rigamonti V, Santambrogio C, Barracchia CG, Munari F, Romeo A, Legname G, Prosperi D, Grandori R, Assfalg M. Dynamic molecular exchange and conformational transitions of alpha-synuclein at the nano-bio interface. Int J Biol Macromol 2020; 154:206-216. [DOI: 10.1016/j.ijbiomac.2020.03.118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 10/24/2022]
|
16
|
Hanif S, Muhammad P, Chesworth R, Rehman FU, Qian RJ, Zheng M, Shi BY. Nanomedicine-based immunotherapy for central nervous system disorders. Acta Pharmacol Sin 2020; 41:936-953. [PMID: 32467570 PMCID: PMC7468531 DOI: 10.1038/s41401-020-0429-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
Central nervous system (CNS) disorders represent a broad spectrum of brain ailments with short- and long-term disabilities, and nanomedicine-based approaches provide a new therapeutic approach to treating CNS disorders. A variety of potential drugs have been discovered to treat several neuronal disorders; however, their therapeutic success can be limited by the presence of the blood-brain barrier (BBB). Furthermore, unique immune functions within the CNS provide novel target mechanisms for the amelioration of CNS diseases. Recently, various therapeutic approaches have been applied to fight brain-related disorders, with moderate outcomes. Among the various therapeutic strategies, nanomedicine-based immunotherapeutic systems represent a new era that can deliver useful cargo with promising pharmacokinetics. These approaches exploit the molecular and cellular targeting of CNS disorders for enhanced safety, efficacy, and specificity. In this review, we focus on the efficacy of nanomedicines that utilize immunotherapy to combat CNS disorders. Furthermore, we detailed summarize nanomedicine-based pathways for CNS ailments that aim to deliver drugs across the BBB by mimicking innate immune actions. Overview of how nanomedicines can utilize multiple immunotherapy pathways to combat CNS disorders. ![]()
Collapse
|
17
|
Jafaripour SS, Gharaghani S, Nazarshodeh E, Haider S, Saboury AA. In silico drug repositioning of FDA-approved drugs to predict new inhibitors for alpha-synuclein aggregation. Comput Biol Chem 2020; 88:107308. [PMID: 32650066 DOI: 10.1016/j.compbiolchem.2020.107308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 05/16/2020] [Accepted: 06/11/2020] [Indexed: 10/24/2022]
Abstract
One of the hallmarks of Parkinson's disease (PD), a long-term neurodegenerative syndrome, is the accumulation of alpha-synuclein (α-syn) fibrils. Despite numerous studies and efforts, inhibition of α-syn protein aggregation is still a challenge. To overcome this issue, we propose an in silico pharmacophore-based repositioning strategy, to find a pharmaceutical drug that, in addition to their defined role, can be used to prevent aggregation of the α-syn protein. Ligand-based pharmacophore modeling was developed and the best model was selected with validation parameters including 72 % sensitivity, 98 % specificity and goodness score about 0.7. The optimal model has three groups of hydrogen bond donor (HBD), three groups of hydrogen bond acceptor (HBA), and two aromatic rings (AR). The FDA-Approved reports in the ZINC15 database were screened with the pharmacophore model taken from inhibitor compounds. The model identified 22 hits, as promising candidate drugs for Parkinson's therapy. It is noteworthy that among these, 10 drugs have been reported to inhibition of α-syn aggregation or treat/reduce Parkinson's pathogenesis. This model was used to virtual screen ZINC, NCI databases, and natural products from the pomegranate. The results of this screen were filtered for their inability to cross the blood-brain barrier, poor oral bioavailability, etc. Finally, the selected compounds of two ZINC and NCI databases were combined and structurally clustered. Remained compounds were clustered in 28 different clusters, and the 17 compounds were introduced as final candidates.
Collapse
Affiliation(s)
| | - Sajjad Gharaghani
- Laboratory of Bioinformatics and Drug Design, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | - Elmira Nazarshodeh
- Laboratory of Bioinformatics and Drug Design, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
18
|
Bartl S, Oueslati A, Southwell AL, Siddu A, Parth M, David LS, Maxan A, Salhat N, Burkert M, Mairhofer A, Friedrich T, Pankevych H, Balazs K, Staffler G, Hayden MR, Cicchetti F, Smrzka OW. Inhibiting cellular uptake of mutant huntingtin using a monoclonal antibody: Implications for the treatment of Huntington's disease. Neurobiol Dis 2020; 141:104943. [PMID: 32407769 DOI: 10.1016/j.nbd.2020.104943] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/08/2020] [Accepted: 05/06/2020] [Indexed: 12/24/2022] Open
Abstract
Huntington's disease (HD) is caused by a highly polymorphic CAG trinucleotide expansion in the gene encoding for the huntingtin protein (HTT). The resulting mutant huntingtin protein (mutHTT) is ubiquitously expressed but also exhibits the ability to propagate from cell-to-cell to disseminate pathology; a property which may serve as a new therapeutic focus. Accordingly, we set out to develop a monoclonal antibody (mAB) targeting a particularly exposed region close to the aa586 caspase-6 cleavage site of the HTT protein. This monoclonal antibody, designated C6-17, effectively binds mutHTT and is able to deplete the protein from cell culture supernatants. Using cell-based assays, we demonstrate that extracellular secretion of mutHTT into cell culture media and its subsequent uptake in recipient HeLa cells can be almost entirely blocked by mAB C6-17. Immunohistochemical stainings of post-mortem HD brain tissue confirmed the specificity of mAB C6-17 to human mutHTT aggregates. These findings demonstrate that mAB C6-17 not only successfully engages with its target, mutHTT, but also inhibits cell uptake suggesting that this antibody could interfere with the pathological processes of mutHTT spreading in vivo.
Collapse
Affiliation(s)
| | - Abid Oueslati
- Université Laval/Centre de recherche du CHU, Québec, Canada
| | | | - Alberto Siddu
- Université Laval/Centre de recherche du CHU, Québec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | - Oskar W Smrzka
- AFFiRiS AG, Vienna, Austria; Ablevia biotech GmbH, Vienna, Austria
| |
Collapse
|
19
|
Qu M. The Neuroprotective Effect of Steroid Receptor Coactivator-Interacting Protein (SIP) in Astrocyte Model of 1-Methyl-4-Phenylpyridinium (MPP⁺)-Induced Parkinson's Disease. Med Sci Monit 2019; 25:5776-5784. [PMID: 31376345 PMCID: PMC6690214 DOI: 10.12659/msm.912106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background The purpose of this study was to investigate the role and mechanism of steroid receptor coactivator-interacting protein (SIP) in an astrocyte model of 1-methyl-4-phenylpyridinium (MPP+)-induced Parkinson’s disease. Material/Methods To perform our study, a Parkinson’s disease cell model was established by treating the rat glioblastoma cell line C6 with MPP+. SIP was overexpressed in C6 cells using SIP-plasmid. Cell viability and apoptosis were analyzed using MTT assay and flow cytometer respectively. Tumor necrosis factor (TNF)-α and interleukin (IL)-1β levels were detected using enzyme linked immunosorbent assay and quantitative reverse transcription PCR. Besides, lactate dehydrogenase (LDH) release, reactive oxygen species (ROS) production, and superoxide dismutase (SOD) enzyme activity were determined in the present study. For protein and mRNA detection, western blot assay, and qRT-PCR were performed respectively. Results SIP was decreased in MPP+-induced C6 cells. SIP overexpression relieved MPP+-induced cytotoxicity of C6 cells, displayed as increased cell viability and reduced cell apoptosis and reduced LDH release. Besides, SIP inhibited MPP+-induced inflammatory response and oxidative stress, evidenced by decreased levels of inflammatory factors (TNF-α and IL-1β), reduced ROS generation and enhanced SOD activity. Moreover, MPP+-induced nuclear factor-κB activation was inhibited by SIP overexpression. Conclusions SIP was downregulated in Parkinson’s disease and it played a protective role in the development Parkinson’s disease, thus may be a promising target for Parkinson’s disease treatment.
Collapse
Affiliation(s)
- Mingwei Qu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei, China (mainland)
| |
Collapse
|
20
|
Spencer B, Trinh I, Rockenstein E, Mante M, Florio J, Adame A, El-Agnaf OMA, Kim C, Masliah E, Rissman RA. Systemic peptide mediated delivery of an siRNA targeting α-syn in the CNS ameliorates the neurodegenerative process in a transgenic model of Lewy body disease. Neurobiol Dis 2019; 127:163-177. [PMID: 30849508 PMCID: PMC6588505 DOI: 10.1016/j.nbd.2019.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/05/2019] [Accepted: 03/04/2019] [Indexed: 02/08/2023] Open
Abstract
Neurodegenerative disorders of the aging population are characterized by progressive accumulation of neuronal proteins such as α-synuclein (α-syn) in Parkinson's Disease (PD) and Amyloid ß (Aß) and Tau in Alzheimer's disease (AD) for which no treatments are currently available. The ability to regulate the expression at the gene transcription level would be beneficial for reducing the accumulation of these proteins or regulating expression levels of other genes in the CNS. Short interfering RNA molecules can bind specifically to target RNAs and deliver them for degradation. This approach has shown promise therapeutically in vitro and in vivo in mouse models of PD and AD and other neurological disorders; however, delivery of the siRNA to the CNS in vivo has been achieved primarily through intra-cerebral or intra-thecal injections that may be less amenable for clinical translation; therefore, alternative approaches for delivery of siRNAs to the brain is needed. Recently, we described a small peptide from the envelope protein of the rabies virus (C2-9r) that was utilized to deliver an siRNA targeting α-syn across the blood brain barrier (BBB) following intravenous injection. This approach showed reduced expression of α-syn and neuroprotection in a toxic mouse model of PD. However, since receptor-mediated delivery is potentially saturable, each allowing the delivery of a limited number of molecules, we identified an alternative peptide for the transport of nucleotides across the BBB based on the apolipoprotein B (apoB) protein targeted to the family of low-density lipoprotein receptors (LDL-R). We used an 11-amino acid sequence from the apoB protein (ApoB11) that, when coupled with a 9-amino acid arginine linker, can transport siRNAs across the BBB to neuronal and glial cells. To examine the value of this peptide mediated oligonucleotide delivery system for PD, we delivered an siRNA targeting the α-syn (siα-syn) in a transgenic mouse model of PD. We found that ApoB11 was effective (comparable to C2-9r) at mediating the delivery of siα-syn into the CNS, co-localized to neurons and glial cells and reduced levels of α-syn protein translation and accumulation. Delivery of ApoB11/siα-syn was accompanied by protection from degeneration of selected neuronal populations in the neocortex, limbic system and striato-nigral system and reduced neuro-inflammation. Taken together, these results suggest that systemic delivery of oligonucleotides targeting α-syn using ApoB11 might be an interesting alternative strategy worth considering for the experimental treatment of synucleinopathies.
Collapse
Affiliation(s)
- Brian Spencer
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Ivy Trinh
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Jazmin Florio
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Omar M A El-Agnaf
- Neurological Disorders Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Changyoun Kim
- Laboratory of Neurogenetics National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Laboratory of Neurogenetics National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Division of Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Veterans Affairs San Diego Healthcare System San Diego, CA, USA.
| |
Collapse
|
21
|
Wentink A, Nussbaum-Krammer C, Bukau B. Modulation of Amyloid States by Molecular Chaperones. Cold Spring Harb Perspect Biol 2019; 11:a033969. [PMID: 30755450 PMCID: PMC6601462 DOI: 10.1101/cshperspect.a033969] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aberrant protein aggregation is a defining feature of most neurodegenerative diseases. During pathological aggregation, key proteins transition from their native state to alternative conformations, which are prone to oligomerize into highly ordered fibrillar states. As part of the cellular quality control machinery, molecular chaperones can intervene at many stages of the aggregation process to inhibit or reverse aberrant protein aggregation or counteract the toxicity associated with amyloid species. Although the action of chaperones is considered cytoprotective, essential housekeeping functions can be hijacked for the propagation and spreading of protein aggregates, suggesting the cellular protein quality control system constitutes a double-edged sword in neurodegeneration. Here, we discuss the various mechanisms used by chaperones to influence protein aggregation into amyloid fibrils to understand how the interplay of these activities produces specific cellular outcomes and to define mechanisms that may be targeted by pharmacological agents for the treatment of neurodegenerative conditions.
Collapse
Affiliation(s)
- Anne Wentink
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| |
Collapse
|
22
|
Sánchez-Vidaña DI, Chow JKW, Hu SQ, Lau BWM, Han YF. Molecular Targets of Bis (7)-Cognitin and Its Relevance in Neurological Disorders: A Systematic Review. Front Neurosci 2019; 13:445. [PMID: 31143096 PMCID: PMC6521802 DOI: 10.3389/fnins.2019.00445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/18/2019] [Indexed: 01/22/2023] Open
Abstract
Background: The exact mechanisms involved in the pathogenesis of neurodegenerative conditions are not fully known. The design of drugs that act on multiple targets represents a promising approach that should be explored for more effective clinical options for neurodegenerative disorders. B7C is s synthetic drug that has been studied for over 20 years and represents a promising multi-target drug for the treatment of neurodegenerative disorders, such as AD. Aims: The present systematic review, thus, aims at examining existing studies on the effect of B7C on different molecular targets and at discussing the relevance of B7C in neurological disorders. Methods: A list of predefined search terms was used to retrieve relevant articles from the databases of Embase, Pubmed, Scopus, and Web of Science. The selection of articles was done by two independent authors, who were considering articles concerned primarily with the evaluation of the effect of B7C on neurological disorders. Only full-text articles written in English were included; whereas, systematic reviews, meta-analyses, book chapters, conference subtracts, and computational studies were excluded. Results: A total of 2,266 articles were retrieved out of which 41 articles were included in the present systematic review. The effect of B7C on molecular targets, including AChE, BChE, BACE-1, NMDA receptor, GABA receptor, NOS, and Kv4.2 potassium channels was evaluated. Moreover, the studies that were included assessed the effect of B7C on biological processes, such as apoptosis, neuritogenesis, and amyloid beta aggregation. The animal studies examined in the review focused on the effect of B7C on cognition and memory. Conclusions: The beneficial effects observed on different molecular targets and biological processes relevant to neurological conditions confirm that B7C is a promising multi-target drug with the potential to treat neurological disorders.
Collapse
Affiliation(s)
| | - Jason Ka Wing Chow
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Sheng Quan Hu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Benson Wui Man Lau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yi-Fan Han
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
23
|
Titze-de-Almeida R, Titze-de-Almeida SS, Ferreira NR, Fontanari C, Faccioli LH, Del Bel E. Suppressing nNOS Enzyme by Small-Interfering RNAs Protects SH-SY5Y Cells and Nigral Dopaminergic Neurons from 6-OHDA Injury. Neurotox Res 2019; 36:117-131. [PMID: 31041676 DOI: 10.1007/s12640-019-00043-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/27/2019] [Accepted: 04/04/2019] [Indexed: 01/11/2023]
Abstract
Nitric oxide (NO) has chemical properties that make it uniquely suitable as an intracellular and intercellular messenger. NO is produced by the activity of the enzyme nitric oxide synthases (NOS). There is substantial and mounting evidence that slight abnormalities of NO may underlie a wide range of neurodegenerative disorders. NO participates of the oxidative stress and inflammatory processes that contribute to the progressive dopaminergic loss in Parkinson's disease (PD). The present study aimed to evaluate in vitro and in vivo the effects of neuronal NOS-targeted siRNAs on the injury caused in dopaminergic neurons by the toxin 6-hidroxydopamine (6-OHDA). First, we confirmed (immunohistochemistry and Western blotting) that SH-SY5Y cell lineage expresses the dopaminergic marker tyrosine hydroxylase (TH) and the protein under analysis, neuronal NOS (nNOS). We designed four siRNAs by using the BIOPREDsi algorithm choosing the one providing the highest knockdown of nNOS mRNA in SH-SY5Y cells, as determined by qPCR. siRNA 4400 carried by liposomes was internalized into cells, caused a concentration-dependent knockdown on nNOS, and reduced the toxicity induced by 6-OHDA (p < 0.05). Regarding in vivo action in the dopamine-depleted animals, intra-striatal injection of siRNA 4400 at 4 days prior 6-OHDA produced a decrease in the rotational behavior induced by apomorphine. Finally, siRNA 4400 mitigated the loss of TH(+) cells in substantia nigra dorsal and ventral part. In conclusion, the suppression of nNOS enzyme by targeted siRNAs modified the progressive death of dopaminergic cells induced by 6-OHDA and merits further pre-clinical investigations as a neuroprotective approach for PD.
Collapse
Affiliation(s)
- Ricardo Titze-de-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, University of Brasília-FAV, Brasília, DF, 70910-900, Brazil
| | - Simoneide S Titze-de-Almeida
- Technology for Gene Therapy Laboratory, Central Institute of Sciences, University of Brasília-FAV, Brasília, DF, 70910-900, Brazil
| | - Nadia Rubia Ferreira
- Department of Basic and Oral Biology, Dental School, University of São Paulo (USP), Ribeirão Preto, SP, 14040-904, Brazil
| | - Caroline Fontanari
- Department of Clinical Analyses, Toxicology and Bromatology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Lúcia Helena Faccioli
- Department of Clinical Analyses, Toxicology and Bromatology, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Elaine Del Bel
- Department of Basic and Oral Biology, Dental School, University of São Paulo (USP), Ribeirão Preto, SP, 14040-904, Brazil.
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo (USP), São Paulo, SP, Brazil.
| |
Collapse
|
24
|
Neuroprotective Effect of Echinacoside in Subacute Mouse Model of Parkinson's Disease. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4379639. [PMID: 30834264 PMCID: PMC6374833 DOI: 10.1155/2019/4379639] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/16/2018] [Accepted: 01/15/2019] [Indexed: 12/20/2022]
Abstract
Objective To study the protective effect of Echinacoside for 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced dopaminergic (DA) neurons injury in the subacute mouse model of Parkinson's disease (PD) and to explore its mechanism of action. Methods We chose 10 weeks of healthy wild type C57BL/6 male mice, hypodermic MPTP 30 mg/kg/day, five days, to prepare PD subacute mouse model. Behavior indexes of open field test and pole test were applied to examine the function of ECH to PD subacute mice model of PD sample action. The effects of ECH on dopaminergic neurons and astrocyte were examined using Immunohistochemistry including tyrosine hydroxylase (TH) and glial fibrillary acidic protein (GFAP) expression. The total numbers of TH-positive neurons and GFAP-positive cells in the substantia nigra pars compacts (SNpc) and ventral tegmental area (VTA) were obtained stereologically using the optical fractionator method. Enzyme-linked immunosorbent assay (ELISA) method was used to detect the inflammatory cytokines in the serum, including TNF-α (Ttumor necrosis factor alpha) and IFN-γ (interferon gamma). Protein expressions of ionized calcium binding adaptor molecule 1 (IBA-1), TNF-α, Cleaved caspase-3, glial derived neurotrophic factor (GDNF), and phosphorylated and total extracellular signal-regulated kinase (p-ERK and ERK) in the anatomical region of substantia nigra (SN) were tested by protein immunoblot method (i.e., Western blotting). Results ECH reversed the reduction of total distance in open field test in MPTP-induced PD model mice (P < 0.01), shortened the return time and total time of PD subacute model mice in pole test (P < 0.01, P < 0.05), significantly reversed the reduction of TH positive neurons induced by MPTP (P < 0.05), and reduced the activation of astrocytes (P < 0.05). Meanwhile, ECH significantly inhibited the expression of IBA-1, Cleaved caspase-3, and TNF-α in midbrain of MPTP model mice (P < 0.05, P < 0.05, and P < 0.05) and upregulated the expression of GDNF (P < 0.05). And ECH lowered the level of TNF-α and IFN-γ in serum (P < 0.05, P < 0.05). Conclusion ECH has protective effects on the MPTP subacute model mice, its mechanism may be through inhibiting activation of microglia and astrocytes, reducing inflammatory reaction and promoting the secretion of neurotrophic factors, and eventually resulting in the reduction of the DA neurons apoptosis.
Collapse
|
25
|
Stephenson D, Hill D, Cedarbaum JM, Tome M, Vamvakas S, Romero K, Conrado DJ, Dexter DT, Seibyl J, Jennings D, Nicholas T, Matthews D, Xie Z, Imam S, Maguire P, Russell D, Gordon MF, Stebbins GT, Somer E, Gallagher J, Roach A, Basseches P, Grosset D, Marek K. The Qualification of an Enrichment Biomarker for Clinical Trials Targeting Early Stages of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2019; 9:553-563. [PMID: 31306141 PMCID: PMC6700608 DOI: 10.3233/jpd-191648] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/31/2019] [Indexed: 12/12/2022]
Abstract
As therapeutic trials target early stages of Parkinson's disease (PD), appropriate patient selection based purely on clinical criteria poses significant challenges. Members of the Critical Path for Parkinson's Consortium formally submitted documentation to the European Medicines Agency (EMA) supporting the use of Dopamine Transporter (DAT) neuroimaging in early PD. Regulatory documents included a comprehensive literature review, a proposed analysis plan of both observational and clinical trial data, and an assessment of biomarker reproducibility and reliability. The research plan included longitudinal analysis of the Parkinson Research Examination of CEP-1347 Trial (PRECEPT) and the Parkinson's Progression Markers Initiative (PPMI) study to estimate the degree of enrichment achieved and impact on future trials in subjects with early motor PD. The presence of reduced striatal DAT binding based on visual reads of single photon emission tomography (SPECT) scans in early motor PD subjects was an independent predictor of faster decline in UPDRS Parts II and III as compared to subjects with scans without evidence of dopaminergic deficit (SWEDD) over 24 months. The EMA issued in 2018 a full Qualification Opinion for the use of DAT as an enrichment biomarker in PD trials targeting subjects with early motor symptoms. Exclusion of SWEDD subjects in future clinical trials targeting early motor PD subjects aims to enrich clinical trial populations with idiopathic PD patients, improve statistical power, and exclude subjects who are unlikely to progress clinically from being exposed to novel test therapeutics.
Collapse
Affiliation(s)
| | | | | | - Maria Tome
- European Medicines Agency, Amsterdam, Netherlands
| | | | | | | | | | - John Seibyl
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | | | | | | | | | - Syed Imam
- U.S. Food and Drug Administration, National Center for Toxicological Research, Jefferson, AR, USA
| | | | - David Russell
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | | | | | | | | | | | | | | | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
| | - on behalf of the Critical Path for Parkinson’s Consortium
- Critical Path Institute, Tucson, AZ, USA
- University College London, UK
- Biogen, Cambridge, MA, USA
- European Medicines Agency, Amsterdam, Netherlands
- Parkinson’s UK, London, UK
- Institute for Neurodegenerative Disorders, New Haven, CT, USA
- Denali Therapeutics, San Francisco, CA, USA
- Pfizer, Groton, CT, USA
- ADM Diagnostics, Northbrook, IL, USA
- UCB, Brussels, Belgium
- CPP Scientific Advisor, PA, USA
- GE Healthcare, London, UK
- Merck & Co., Philadelphia, PA, USA
- University of Glasgow, Scotland
- Rush University, Chicago, IL, USA
- U.S. Food and Drug Administration, National Center for Toxicological Research, Jefferson, AR, USA
| |
Collapse
|
26
|
Signature of Aberrantly Expressed microRNAs in the Striatum of Rotenone-Induced Parkinsonian Rats. Neurochem Res 2018; 43:2132-2140. [DOI: 10.1007/s11064-018-2638-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/25/2018] [Accepted: 09/11/2018] [Indexed: 12/21/2022]
|
27
|
Du RH, Zhou Y, Xia ML, Lu M, Ding JH, Hu G. α-Synuclein disrupts the anti-inflammatory role of Drd2 via interfering β-arrestin2-TAB1 interaction in astrocytes. J Neuroinflammation 2018; 15:258. [PMID: 30200997 PMCID: PMC6131810 DOI: 10.1186/s12974-018-1302-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/31/2018] [Indexed: 12/13/2022] Open
Abstract
Background α-Synuclein (α-Syn)-induced neuroinflammation plays a crucial role in the pathogenesis of Parkinson’s disease (PD). Dopamine D2 receptor (Drd2) has been regarded as a potential anti-inflammatory target in the therapy of neurodegenerative diseases. However, the effect of astrocytic Drd2 in α-Syn-induced neuroinflammation remains unclear. Methods The effect of Drd2 on neuroinflammation was examined in mouse primary astrocyte in vitro and A53T transgenic mice in vivo. The inflammatory responses of astrocyte were detected using immunofluorescence, ELISA, and qRT-PCR. The details of molecular mechanism were assessed using Western blotting and protein-protein interaction assays. Results We showed that the selective Drd2 agonist quinpirole suppressed inflammation in the midbrain of wild-type mice, but not in α-Syn-overexpressed mice. We also found that Drd2 agonists significantly alleviated LPS-induced inflammatory response in astrocytes, but failed to suppress α-Syn-induced inflammatory response. The anti-inflammation effect of Drd2 was dependent on β-arrestin2-mediated signaling, but not classical G protein pathway. α-Syn reduced the expression of β-arrestin2 in astrocytes. Increased the β-arrestin2 expression restored in the anti-inflammation of Drd2 in α-Syn-induced inflammation. Furthermore, we demonstrated that α-Syn disrupted the anti-inflammation of Drd2 via inhibiting the association of β-arrestin2 with transforming growth factor-beta-activated kinase 1 (TAK1)-binding protein 1 (TAB1) and promoting TAK1-TAB1 interaction in astrocytes. Conclusions Our study illustrates that astrocytic Drd2 inhibits neuroinflammation through a β-arrestin2-dependent mechanism and provides a new strategy for treatment of PD. Our findings also reveal that α-Syn disrupts the function of β-arrestin2 and inflammatory pathways in the pathogenesis of PD.
Collapse
Affiliation(s)
- Ren-Hong Du
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Yan Zhou
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China.,Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Mei-Ling Xia
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Ming Lu
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Jian-Hua Ding
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Gang Hu
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Nongmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China. .,Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, People's Republic of China.
| |
Collapse
|
28
|
Zhu J, Hu Z, Han X, Wang D, Jiang Q, Ding J, Xiao M, Wang C, Lu M, Hu G. Dopamine D2 receptor restricts astrocytic NLRP3 inflammasome activation via enhancing the interaction of β-arrestin2 and NLRP3. Cell Death Differ 2018; 25:2037-2049. [PMID: 29786071 DOI: 10.1038/s41418-018-0127-2] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/08/2018] [Accepted: 05/02/2018] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are involved in the neuroinflammation of neurodegenerative diseases, such as Parkinson's disease (PD). Among the numerous inflammatory cytokines, interleukin-1β (IL-1β) produced by astrocytic Nod-like receptor protein (NLRP) inflammasome is crucial in the pathogenesis of PD. β-arrestin2-mediated dopamine D2 receptor (Drd2) signal transduction has been regarded as a potential anti-inflammatory target. Our previous study revealed that astrocytic Drd2 suppresses neuroinflammation in the central nervous system. However, the role of Drd2 in astrocytic NLRP3 inflammasome activation and subsequent IL-1β production remains unclear. In the present study, we used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced PD mouse model to investigate whether Drd2 could suppress astrocytic NLRP3 inflammasome activation. We showed that Drd2 agonist inhibited NLRP3 inflammasome activation, evidenced by decreased caspase-1 expression and reduced IL-1β release in the midbrain of wild type mice. The anti-inflammasome effect of Drd2 was abolished in β-arrestin2 knockout and β-arrestin2 small interfering RNA-injected mice, suggesting a critical role of β-arrestin2 in Drd2-regulated NLRP3 inflammasome activation. We also found that Drd2 agonists suppressed the upregulation of caspase-1 and IL-1β expression in primary cultured mouse astrocytes in response to the activation of NLRP3 inflammasome induced by lipopolysaccharide plus adenosine triphosphate. Furthermore, we demonstrated that β-arrestin2 mediated the inhibitory effect of Drd2 on NLRP3 inflammasome activation via interacting with NLRP3 and interfering the inflammasome assembly. Collectively, our study illustrates that astrocytic Drd2 inhibits NLRP3 inflammasome activation through a β-arrestin2-dependent mechanism, and provides a new strategy for treatment of PD.
Collapse
Affiliation(s)
- Jialei Zhu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Zhaoli Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Xiaojuan Han
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Dongshuo Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China
| | - Qingling Jiang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Cong Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China.
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China. .,Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
29
|
Shvadchak VV, Afitska K, Yushchenko DA. Inhibition of α-Synuclein Amyloid Fibril Elongation by Blocking Fibril Ends. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201801071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Volodymyr V. Shvadchak
- Laboratory of Chemical Biology; Institute of Organic Chemistry and Biochemistry; Academy of Sciences of the Czech Republic; Flemingovo nam. 2 16610 Prague 6 Czech Republic
| | - Kseniia Afitska
- Laboratory of Chemical Biology; Institute of Organic Chemistry and Biochemistry; Academy of Sciences of the Czech Republic; Flemingovo nam. 2 16610 Prague 6 Czech Republic
| | - Dmytro A. Yushchenko
- Laboratory of Chemical Biology; Institute of Organic Chemistry and Biochemistry; Academy of Sciences of the Czech Republic; Flemingovo nam. 2 16610 Prague 6 Czech Republic
- Cell Biology & Biophysics Unit; European Molecular Biology Laboratory (EMBL); Meyerhofstraße 1 69117 Heidelberg Germany
| |
Collapse
|
30
|
Shvadchak VV, Afitska K, Yushchenko DA. Inhibition of α-Synuclein Amyloid Fibril Elongation by Blocking Fibril Ends. Angew Chem Int Ed Engl 2018; 57:5690-5694. [DOI: 10.1002/anie.201801071] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/04/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Volodymyr V. Shvadchak
- Laboratory of Chemical Biology; Institute of Organic Chemistry and Biochemistry; Academy of Sciences of the Czech Republic; Flemingovo nam. 2 16610 Prague 6 Czech Republic
| | - Kseniia Afitska
- Laboratory of Chemical Biology; Institute of Organic Chemistry and Biochemistry; Academy of Sciences of the Czech Republic; Flemingovo nam. 2 16610 Prague 6 Czech Republic
| | - Dmytro A. Yushchenko
- Laboratory of Chemical Biology; Institute of Organic Chemistry and Biochemistry; Academy of Sciences of the Czech Republic; Flemingovo nam. 2 16610 Prague 6 Czech Republic
- Cell Biology & Biophysics Unit; European Molecular Biology Laboratory (EMBL); Meyerhofstraße 1 69117 Heidelberg Germany
| |
Collapse
|
31
|
Zhang J, Culp ML, Craver JG, Darley-Usmar V. Mitochondrial function and autophagy: integrating proteotoxic, redox, and metabolic stress in Parkinson's disease. J Neurochem 2018; 144:691-709. [PMID: 29341130 PMCID: PMC5897151 DOI: 10.1111/jnc.14308] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/04/2018] [Accepted: 01/09/2018] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a movement disorder with widespread neurodegeneration in the brain. Significant oxidative, reductive, metabolic, and proteotoxic alterations have been observed in PD postmortem brains. The alterations of mitochondrial function resulting in decreased bioenergetic health is important and needs to be further examined to help develop biomarkers for PD severity and prognosis. It is now becoming clear that multiple hits on metabolic and signaling pathways are likely to exacerbate PD pathogenesis. Indeed, data obtained from genetic and genome association studies have implicated interactive contributions of genes controlling protein quality control and metabolism. For example, loss of key proteins that are responsible for clearance of dysfunctional mitochondria through a process called mitophagy has been found to cause PD, and a significant proportion of genes associated with PD encode proteins involved in the autophagy-lysosomal pathway. In this review, we highlight the evidence for the targeting of mitochondria by proteotoxic, redox and metabolic stress, and the role autophagic surveillance in maintenance of mitochondrial quality. Furthermore, we summarize the role of α-synuclein, leucine-rich repeat kinase 2, and tau in modulating mitochondrial function and autophagy. Among the stressors that can overwhelm the mitochondrial quality control mechanisms, we will discuss 4-hydroxynonenal and nitric oxide. The impact of autophagy is context depend and as such can have both beneficial and detrimental effects. Furthermore, we highlight the potential of targeting mitochondria and autophagic function as an integrated therapeutic strategy and the emerging contribution of the microbiome to PD susceptibility.
Collapse
Affiliation(s)
- Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
- Department of Veterans Affairs, Birmingham VA Medical Center
| | - M Lillian Culp
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| | - Jason G Craver
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| | - Victor Darley-Usmar
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| |
Collapse
|
32
|
Rockenstein E, Ostroff G, Dikengil F, Rus F, Mante M, Florio J, Adame A, Trinh I, Kim C, Overk C, Masliah E, Rissman RA. Combined Active Humoral and Cellular Immunization Approaches for the Treatment of Synucleinopathies. J Neurosci 2018; 38:1000-1014. [PMID: 29246926 PMCID: PMC5783958 DOI: 10.1523/jneurosci.1170-17.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/30/2022] Open
Abstract
Dementia with Lewy bodies, Parkinson's disease, and Multiple System Atrophy are age-related neurodegenerative disorders characterized by progressive accumulation of α-synuclein (α-syn) and jointly termed synucleinopathies. Currently, no disease-modifying treatments are available for these disorders. Previous preclinical studies demonstrate that active and passive immunizations targeting α-syn partially ameliorate behavioral deficits and α-syn accumulation; however, it is unknown whether combining humoral and cellular immunization might act synergistically to reduce inflammation and improve microglial-mediated α-syn clearance. Since combined delivery of antigen plus rapamycin (RAP) in nanoparticles is known to induce antigen-specific regulatory T cells (Tregs), we adapted this approach to α-syn using the antigen-presenting cell-targeting glucan microparticle (GP) vaccine delivery system. PDGF-α-syn transgenic (tg) male and female mice were immunized with GP-alone, GP-α-syn (active humoral immunization), GP+RAP, or GP+RAP/α-syn (combined active humoral and Treg) and analyzed using neuropathological and biochemical markers. Active immunization resulted in higher serological total IgG, IgG1, and IgG2a anti-α-syn levels. Compared with mice immunized with GP-alone or GP-α-syn, mice vaccinated with GP+RAP or GP+RAP/α-syn displayed increased numbers of CD25-, FoxP3-, and CD4-positive cells in the CNS. GP-α-syn or GP+RAP/α-syn immunizations resulted in a 30-45% reduction in α-syn accumulation, neuroinflammation, and neurodegeneration. Mice immunized with GP+RAP/α-syn further rescued neurons and reduced neuroinflammation. Levels of TGF-β1 were increased with GP+RAP/α-syn immunization, while levels of TNF-α and IL-6 were reduced. We conclude that the observed effects of GP+RAP/α-syn immunization support the hypothesis that cellular immunization may enhance the effects of active immunotherapy for the treatment of synucleinopathies.SIGNIFICANCE STATEMENT We show that a novel vaccination modality combining an antigen-presenting cell-targeting glucan particle (GP) vaccine delivery system with encapsulated antigen (α-synuclein) + rapamycin (RAP) induced both strong anti-α-synuclein antibody titers and regulatory T cells (Tregs). This vaccine, collectively termed GP+RAP/α-syn, is capable of triggering neuroprotective Treg responses in synucleinopathy models, and the combined vaccine is more effective than the humoral or cellular immunization alone. Together, these results support the further development of this multifunctional vaccine approach for the treatment of synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies, and multiple systems atrophy.
Collapse
Affiliation(s)
- Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Gary Ostroff
- University of Mass Massachusetts Medical School, Program in Molecular Medicine Worcester, Massachusetts 01605
| | - Fusun Dikengil
- University of Mass Massachusetts Medical School, Program in Molecular Medicine Worcester, Massachusetts 01605
| | - Florentina Rus
- University of Mass Massachusetts Medical School, Program in Molecular Medicine Worcester, Massachusetts 01605
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Jazmin Florio
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Ivy Trinh
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Changyoun Kim
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Cassia Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624,
- Veterans Affairs San Diego Healthcare System, La Jolla, California 92161
| |
Collapse
|
33
|
Overk C, Rockenstein E, Valera E, Stefanova N, Wenning G, Masliah E. Multiple system atrophy: experimental models and reality. Acta Neuropathol 2018; 135:33-47. [PMID: 29058121 PMCID: PMC6156777 DOI: 10.1007/s00401-017-1772-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/29/2017] [Accepted: 09/29/2017] [Indexed: 01/08/2023]
Abstract
Multiple system atrophy (MSA) is a rapidly progressing fatal synucleinopathy of the aging population characterized by parkinsonism, dysautonomia, and in some cases ataxia. Unlike other synucleinopathies, in this disorder the synaptic protein, α-synuclein (α-syn), predominantly accumulates in oligodendroglial cells (and to some extent in neurons), leading to maturation defects of oligodendrocytes, demyelination, and neurodegeneration. The mechanisms through which α-syn deposits occur in oligodendrocytes and neurons in MSA are not completely clear. While some studies suggest that α-syn might transfer from neurons to glial cells, others propose that α-syn might be aberrantly overexpressed by oligodendroglial cells. A number of in vivo models have been developed, including transgenic mice overexpressing α-syn under oligodendroglial promoters (e.g.: MBP, PLP, and CNP). Other models have been recently developed either by injecting synthetic α-syn fibrils or brain homogenates from patients with MSA into wild-type mice or by using viral vectors expressing α-syn under the MBP promoter in rats and non-human primates. Each of these models reproduces some of the neuropathological and functional aspects of MSA; however, none of them fully replicate the spectrum of MSA. Understanding better the mechanisms of how α-syn accumulates in oligodendrocytes and neurons will help in developing better models that recapitulate various pathogenic aspects of MSA in combination with translatable biomarkers of early stages of the disease that are necessary to devise disease-modifying therapeutics for MSA.
Collapse
Affiliation(s)
- Cassia Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093-0624, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093-0624, USA
| | - Elvira Valera
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093-0624, USA
| | - Nadia Stefanova
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor Wenning
- Division of Clinical Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093-0624, USA.
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
34
|
Guan J, Yang B, Fan Y, Zhang J. GPER Agonist G1 Attenuates Neuroinflammation and Dopaminergic Neurodegeneration in Parkinson Disease. Neuroimmunomodulation 2017; 24:60-66. [PMID: 28810246 DOI: 10.1159/000478908] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 06/22/2017] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE Epidemiological studies have shown that women of reproductive age have much less possibility of developing Parkinson disease (PD) than men. The beneficial effect of estrogen also has been well-described in both culture and animal models of PD. G protein-coupled estrogen receptor (GPER) is a membrane-associated estrogen receptor, and displayed a neuroprotective role in a mouse model of PD. Since GPER is highly expressed in microglia, we speculate that GPER mediates the neuroprotective function of estradiol through suppressing the neuroinflammation of PD. METHODS We investigated the effects of GPER agonist G1 and GPER antagonist G15 on the neurodegeneration of dopaminergic neuron, the activation of microglia, and the production of IL-1β, TNF-α, and IL-6 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced animal model of parkinsonism. Furthermore, we confirmed the effects of GPER activation on the production of IL-1β, TNF-α, and IL-6 in an in vitro MPP+ model in BV2 microglial cells. RESULTS After 12-day treatment with G1, mice showed an increase in the number of tyrosine hydroxylase-immunoreactive cells, reduced activation of microglia, and the abatement of proinflammatory cytokines, and the anti-inflammatory effect of G1 was abolished by G15. Meanwhile, in vitro studies demonstrated that GPER activation also reduced the release of proinflammatory cytokines from BV2 microglial cells after MPP+ stimulation. CONCLUSION Our data suggest that GPER mediates the anti-neuroinflammatory effect of estrogen in experimental PD progression.
Collapse
Affiliation(s)
- Jing Guan
- State Key Laboratory of Reproductive Medicine, Nanjing Maternity and Child Health Care Hospital, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | | | | | | |
Collapse
|
35
|
Lindenbach D, Das B, Conti MM, Meadows SM, Dutta AK, Bishop C. D-512, a novel dopamine D 2/3 receptor agonist, demonstrates greater anti-Parkinsonian efficacy than ropinirole in Parkinsonian rats. Br J Pharmacol 2017; 174:3058-3071. [PMID: 28667675 DOI: 10.1111/bph.13937] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/24/2017] [Accepted: 06/19/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Symptoms of Parkinson's disease are commonly managed using selective dopamine D2/3 receptor agonists, including ropinirole. While D2/3 agonists are useful in early-stage Parkinson's disease, they tend to lose efficacy in later disease stages and do not appear to modify disease progression. We have recently developed a novel 'multifunctional' compound, D-512: a high-affinity D2/3 receptor agonist with antioxidant and other neuroprotective properties that may limit Parkinson's disease progression. This study sought to compare the anti-Parkinsonian properties of the clinically used compound, ropinirole, with those of the novel compound, D-512. EXPERIMENTAL APPROACH A rat model of Parkinson's disease was created by unilaterally infusing 6-hydroxydopamine, a dopamine neurotoxin, into the medial forebrain bundle. D-512 was compared with ropinirole for ability to stimulate spontaneous motor activity and reverse Parkinsonian akinesia. These beneficial effects were compared against each drug's liability to provoke dyskinesia, a common motor side effect. KEY RESULTS Both compounds increased spontaneous movement, but D-512 showed a longer duration of action. Only D-512 was able to significantly reverse forelimb akinesia. Drug-induced dyskinesia was similar for equivalent doses. CONCLUSIONS AND IMPLICATIONS Compared with ropinirole, D-512 showed greater peak-dose efficacy and a longer duration of action, despite a similar side-effect profile. Our results add to earlier data showing that D-512 is superior to available D2/3 agonists and could merit clinical investigation.
Collapse
Affiliation(s)
- David Lindenbach
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University - State University of New York, Binghamton, NY, USA
| | - Banibrata Das
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Melissa M Conti
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University - State University of New York, Binghamton, NY, USA
| | - Samantha M Meadows
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University - State University of New York, Binghamton, NY, USA
| | - Aloke K Dutta
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Christopher Bishop
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University - State University of New York, Binghamton, NY, USA
| |
Collapse
|
36
|
Castro Caldas A, Levin J, Djaldetti R, Rascol O, Wenning G, Ferreira JJ. Critical appraisal of clinical trials in multiple system atrophy: Toward better quality. Mov Disord 2017; 32:1356-1364. [PMID: 28782838 DOI: 10.1002/mds.27080] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/02/2017] [Accepted: 05/19/2017] [Indexed: 01/20/2023] Open
Abstract
Multiple system atrophy (MSA) is a rare neurodegenerative disease of undetermined cause. Although many clinical trials have been conducted, there is still no treatment that cures the disease or slows its progression. We sought to assess the clinical trials, methodology, and quality of reporting of clinical trails conducted in MSA patients. We conducted a systematic review of all trials with at least 1 MSA patient subject to any pharmacological/nonpharmacological interventions. Two independent reviewers evaluated the methodological characteristics and quality of reporting of trials. A total of 60 clinical trials were identified, including 1375 MSA patients. Of the trials, 51% (n = 31) were single-arm studies. A total of 28% (n = 17) had a parallel design, half of which (n = 13) were placebo controlled. Of the studies, 8 (13.3%) were conducted in a multicenter setting, 3 of which were responsible for 49.3% (n = 678) of the total included MSA patients. The description of primary outcomes was unclear in 60% (n = 40) of trials. Only 10 (16.7%) clinical trials clearly described the randomization process. Blinding of the participants, personnel, and outcome assessments were at high risk of bias in the majority of studies. The number of dropouts/withdrawals was high (n = 326, 23.4% among the included patients). Overall, the design and quality of reporting of the reviewed studies is unsatisfactory. The most frequent clinical trials were small and single centered. Inadequate reporting was related to the information on the randomization process, sequence generation, allocation concealment, blinding of participants, and sample size calculations. Although improved during the recent years, methodological quality and trial design need to be optimized to generate more informative results. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ana Castro Caldas
- Neurology Service, Department of Neurosciences, Hospital de Santa Maria, Lisbon, Portugal.,Clinical Pharmacology Unit, Instituto de Medicina Molecular, Lisbon, Portugal.,Campus Neurológico Sénior, Torres Vedras, Portugal
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany
| | - Ruth Djaldetti
- Department of Neurology, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel
| | - Olivier Rascol
- Department of Neurosciences and Clinical Pharmacology, Reference Center for MSA, Clinical Investigation Center (CIC) 1436, and NeuroToul Center of Excellence in Neurodegeneration (COEN) Center, University of Toulouse, Institut national de la santé et de la recherche médicale (INSERM), University Hospital of Toulouse, Toulouse, France
| | - Gregor Wenning
- Department of Neurology, University Hospital, Innsbruck, Austria
| | - Joaquim J Ferreira
- Clinical Pharmacology Unit, Instituto de Medicina Molecular, Lisbon, Portugal.,Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | | |
Collapse
|
37
|
Chiti F, Dobson CM. Protein Misfolding, Amyloid Formation, and Human Disease: A Summary of Progress Over the Last Decade. Annu Rev Biochem 2017; 86:27-68. [DOI: 10.1146/annurev-biochem-061516-045115] [Citation(s) in RCA: 1714] [Impact Index Per Article: 244.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Peptides and proteins have been found to possess an inherent tendency to convert from their native functional states into intractable amyloid aggregates. This phenomenon is associated with a range of increasingly common human disorders, including Alzheimer and Parkinson diseases, type II diabetes, and a number of systemic amyloidoses. In this review, we describe this field of science with particular reference to the advances that have been made over the last decade in our understanding of its fundamental nature and consequences. We list the proteins that are known to be deposited as amyloid or other types of aggregates in human tissues and the disorders with which they are associated, as well as the proteins that exploit the amyloid motif to play specific functional roles in humans. In addition, we summarize the genetic factors that have provided insight into the mechanisms of disease onset. We describe recent advances in our knowledge of the structures of amyloid fibrils and their oligomeric precursors and of the mechanisms by which they are formed and proliferate to generate cellular dysfunction. We show evidence that a complex proteostasis network actively combats protein aggregation and that such an efficient system can fail in some circumstances and give rise to disease. Finally, we anticipate the development of novel therapeutic strategies with which to prevent or treat these highly debilitating and currently incurable conditions.
Collapse
Affiliation(s)
- Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” Section of Biochemistry, Università di Firenze, 50134 Firenze, Italy
| | - Christopher M. Dobson
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
38
|
A natural product from Cannabis sativa subsp. sativa inhibits homeodomain-interacting protein kinase 2 (HIPK2), attenuating MPP + -induced apoptosis in human neuroblastoma SH-SY5Y cells. Bioorg Chem 2017; 72:64-73. [DOI: 10.1016/j.bioorg.2017.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 03/09/2017] [Accepted: 03/21/2017] [Indexed: 12/27/2022]
|
39
|
El-Agnaf O, Overk C, Rockenstein E, Mante M, Florio J, Adame A, Vaikath N, Majbour N, Lee SJ, Kim C, Masliah E, Rissman RA. Differential effects of immunotherapy with antibodies targeting α-synuclein oligomers and fibrils in a transgenic model of synucleinopathy. Neurobiol Dis 2017; 104:85-96. [PMID: 28476636 DOI: 10.1016/j.nbd.2017.05.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 11/17/2022] Open
Abstract
Disorders with progressive accumulation of α-synuclein (α-syn) are a common cause of dementia and parkinsonism in the aging population. Accumulation and propagation of α-syn play a role in the pathogenesis of these disorders. Previous studies have shown that immunization with antibodies that recognize C-terminus of α-syn reduces the intra-neuronal accumulation of α-syn and related deficits in transgenic models of synucleinopathy. These studies employed antibodies that recognize epitopes within monomeric and aggregated α-syn that were generated through active immunization or administered via passive immunization. However, it is possible that more specific effects might be achieved with antibodies recognizing selective species of the α-syn aggregates. In this respect we recently developed antibodies that differentially recognized various oligomers (Syn-O1, -O2, and -O4) and fibrilar (Syn-F1 and -F2) forms of α-syn. For this purpose wild-type α-syn transgenic (line 61) mice were immunized with these 5 different antibodies and neuropathologically and biochemically analyzed to determine which was most effective at reducing α-syn accumulation and related deficits. We found that Syn-O1, -O4 and -F1 antibodies were most effective at reducing accumulation of α-syn oligomers in multiple brain regions and at preventing neurodegeneration. Together this study supports the notion that selective antibodies against α-syn might be suitable for development new treatments for synucleinopathies such as PD and DLB.
Collapse
Affiliation(s)
- Omar El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 5825, Doha, Qatar; Life Sciences Division, College of Science and Engineering, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, PO Box 5825, Doha, Qatar
| | - Cassia Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Jazmin Florio
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Nishant Vaikath
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 5825, Doha, Qatar
| | - Nour Majbour
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 5825, Doha, Qatar
| | - Seung-Jae Lee
- Department of Biomedical Sciences and Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Changyoun Kim
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States; Department of Pathology, University of California, San Diego, La Jolla, CA 92093, United States
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States; Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, United States.
| |
Collapse
|
40
|
Giffin JC, Richards RC, Craft C, Jahan N, Leggiadro C, Chopin T, Szemerda M, MacKinnon SL, Ewart KV. An extract of the marine alga Alaria esculenta modulates α-synuclein folding and amyloid formation. Neurosci Lett 2017; 644:87-93. [PMID: 28237800 DOI: 10.1016/j.neulet.2017.02.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 02/06/2017] [Accepted: 02/21/2017] [Indexed: 10/20/2022]
Abstract
The conversion of α-synuclein from its natively unfolded and α-helical tetrameric forms to an amyloid conformation is central to the emergence of Parkinson's disease. Therefore, prevention of this conversion may offer an effective way of avoiding the onset of this disease or delaying its progress. At different concentrations, an aqueous extract from the edible winged kelp (Alaria esculenta), was shown to lower and to raise the melting point of α-synuclein. Size fractionation of the extract resulted in the separation of these distinct activities. The fraction below 5kDa decreased the melting point of α-synuclein, whereas the fraction above 10kDa raised the melting point. Both of these fractions were found to inhibit the formation of amyloid aggregates by α-synuclein, measured by thioflavin T dye-binding assays; this effect was further confirmed by transmission electron microscopy showing the inhibition of fibril formation. Circular dichroism analysis suggested that the incubation of α-synuclein under fibrillation conditions resulted in the loss of substantial native helical structure in the presence and absence of the fractions. It is therefore likely that the fractions inhibit fibrillation by interacting with the unfolded form of α-synuclein.
Collapse
Affiliation(s)
- James C Giffin
- Department of Biology, Dalhousie University, P.O. Box 15000, Halifax, NS B3H 4R2, Canada
| | - Robert C Richards
- Aquatic and Crop Resource Development, National Research Council,Sandy Cove Road, Ketch Harbour, NS B3 V 1K9, Canada
| | - Cheryl Craft
- Aquatic and Crop Resource Development, National Research Council,1411 Oxford St., Halifax, NS B3H 3Z1, Canada
| | - Nusrat Jahan
- Aquatic and Crop Resource Development, National Research Council,1411 Oxford St., Halifax, NS B3H 3Z1, Canada
| | - Cindy Leggiadro
- Aquatic and Crop Resource Development, National Research Council,Sandy Cove Road, Ketch Harbour, NS B3 V 1K9, Canada
| | - Thierry Chopin
- Canadian Integrated Multi-Trophic Aquaculture Network, University of New Brunswick, Saint John, NB E2L 4L5, Canada
| | - Michael Szemerda
- Cooke Aquaculture Inc., 874 Main St, Blacks Harbour, NB E5H 1E6, Canada
| | - Shawna L MacKinnon
- Aquatic and Crop Resource Development, National Research Council,1411 Oxford St., Halifax, NS B3H 3Z1, Canada
| | - K Vanya Ewart
- Department of Biology, Dalhousie University, P.O. Box 15000, Halifax, NS B3H 4R2, Canada; Department of Biochemistry and Molecular Biology, Dalhousie University, P.O. Box 15000, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
41
|
Schulz JB, Hausmann L, Hardy J. 199 years of Parkinson disease - what have we learned and what is the path to the future? J Neurochem 2016; 139 Suppl 1:3-7. [PMID: 27581372 DOI: 10.1111/jnc.13733] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 12/21/2022]
Abstract
In 1817, 199 years ago, James Parkinson described for the first time in 'An Essay on the Shaking Palsy' the symptoms of the disease that was later named Parkinson Disease. The current special issue of the Journal of Neurochemistry is dedicated to the discoveries and advances that have been made since, leading to a better understanding of this neurodegenerative disease and of potential treatment options. Reputed researchers cover various aspects from neuroanatomical basics; genetic and molecular risk factors such as LRRK2; the available cell and animal models that mimic crucial features of the pathophysiology; to clinical aspects and treatments, including deep brain stimulation. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Jörg B Schulz
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany. .,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany.
| | - Laura Hausmann
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany
| | - John Hardy
- Reta Lila Weston Research Laboratories, UCL Institute of Neurology, Queen Square, London, WC1N3BG, UK
| |
Collapse
|
42
|
Larrick JW, Alfenito MR, Scott JK, Parren PWHI, Burton DR, Bradbury ARM, Lemere CA, Messer A, Huston JS, Carter PJ, Veldman T, Chester KA, Schuurman J, Adams GP, Reichert JM. Antibody Engineering & Therapeutics 2016: The Antibody Society's annual meeting, December 11-15, 2016, San Diego, CA. MAbs 2016; 8:1425-1434. [PMID: 27557809 PMCID: PMC5098447 DOI: 10.1080/19420862.2016.1227665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Antibody Engineering & Therapeutics, the largest meeting devoted to antibody science and technology and the annual meeting of The Antibody Society, will be held in San Diego, CA on December 11-15, 2016. Each of 14 sessions will include six presentations by leading industry and academic experts. In this meeting preview, the session chairs discuss the relevance of their topics to current and future antibody therapeutics development. Session topics include bispecifics and designer polyclonal antibodies; antibodies for neurodegenerative diseases; the interface between passive and active immunotherapy; antibodies for non-cancer indications; novel antibody display, selection and screening technologies; novel checkpoint modulators / immuno-oncology; engineering antibodies for T-cell therapy; novel engineering strategies to enhance antibody functions; and the biological Impact of Fc receptor engagement. The meeting will open with keynote speakers Dennis R. Burton (The Scripps Research Institute), who will review progress toward a neutralizing antibody-based HIV vaccine; Olivera J. Finn, (University of Pittsburgh School of Medicine), who will discuss prophylactic cancer vaccines as a source of therapeutic antibodies; and Paul Richardson (Dana-Farber Cancer Institute), who will provide a clinical update on daratumumab for multiple myeloma. In a featured presentation, a representative of the World Health Organization's INN expert group will provide a perspective on antibody naming. “Antibodies to watch in 2017” and progress on The Antibody Society's 2016 initiatives will be presented during the Society's special session. In addition, two pre-conference workshops covering ways to accelerate antibody drugs to the clinic and the applications of next-generation sequencing in antibody discovery and engineering will be held on Sunday December 11, 2016.
Collapse
Affiliation(s)
- James W Larrick
- a Panorama Research Institute and Velocity Pharmaceutical Development , South San Francisco , CA , USA
| | | | | | - Paul W H I Parren
- d Genmab , Utrecht , the Netherlands.,e Leiden University Medical Center , Leiden , the Netherlands
| | | | | | - Cynthia A Lemere
- h Brigham and Woman's Hospital and Harvard Medical School , Boston MA , USA
| | - Anne Messer
- i Regenerative Research Foundation , Rensselaer , NY , USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Guo YJ, Dong SY, Cui XX, Feng Y, Liu T, Yin M, Kuo SH, Tan EK, Zhao WJ, Wu YC. Resveratrol alleviates MPTP-induced motor impairments and pathological changes by autophagic degradation of α-synuclein via SIRT1-deacetylated LC3. Mol Nutr Food Res 2016; 60:2161-2175. [PMID: 27296520 DOI: 10.1002/mnfr.201600111] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/07/2016] [Accepted: 05/30/2016] [Indexed: 12/21/2022]
Abstract
SCOPE The accumulation of misfolded α-synuclein in dopaminergic neurons is the leading cause of Parkinson's disease (PD). Resveratrol (RV), a polyphenolic compound derived from grapes and red wine, exerts a wide range of beneficial effects via activation of sirtuin 1 (SIRT1) and induction of vitagenes. Here, we assessed the role of RV in a 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) induced mouse model of PD and explored its potential mechanisms. METHODS AND RESULTS RV and EX527, a specific inhibitor of SIRT1, were administered before and after MPTP treatment. RV protected against MPTP-induced loss of dopaminergic neurons, and decreases in tyrosine hydroxylase and dopamine levels, as well as behavioral impairments. Meanwhile, RV administration activated SIRT1. Microtubule-associated protein 1 light chain 3 (LC3) was then deacetylated and redistributed from the nucleus to the cytoplasm, which provoked the autophagic degradation of α-synuclein in dopaminergic neurons. Furthermore, EX527 antagonized the neuroprotective effects of RV by reducing LC3 deacetylation and subsequent autophagic degradation of α-synuclein. CONCLUSION We showed that RV ameliorated both motor deficits and pathological changes in MPTP-treated mice via activation of SIRT1 and subsequent LC3 deacetylation-mediated autophagic degradation of α-synuclein. Our observations suggest that RV may be a potential prophylactic and/or therapeutic agent for PD.
Collapse
Affiliation(s)
- Yan-Jie Guo
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Su-Yan Dong
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Xin-Xin Cui
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Ya Feng
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Longhua Hospital, , Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Ming Yin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Sheng-Han Kuo
- Department of Neurology, College of Physicians and Surgeons, Columbia University, NY, USA
| | - Eng-King Tan
- Department of Neurology, Singapore General Hospital, National Neuroscience Institute, Singapore
| | - Wen-Juan Zhao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Yun-Cheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China.
| |
Collapse
|