1
|
Li J, Liu H, Hu X, Zhang S, Yu Q, Kuang G, Liu L, Yu D, Huang J, Xia Y, Wang T, Xiong N. NR1H4 ameliorates Parkinson's disease via inhibiting astrocyte activation and neuroinflammation in a CEBPβ/NF-κB dependent manner. Int Immunopharmacol 2024; 142:113087. [PMID: 39241522 DOI: 10.1016/j.intimp.2024.113087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Parkinson's Disease (PD) is a degenerative disease driven by neuroinflammation. Nuclear receptor subfamily 1 group H member 4 (NR1H4), a nuclear receptor involved in metabolic and inflammatory regulation, is found to be widely expressed in central nervous system. Previous studies suggested the protective role of NR1H4 in various diseases related to inflammation, whether NR1H4 participates in PD progression remains unknown. To investigate the role of NR1H4 in neuroinflammation regulation, especially astrocyte activation during PD, siRNA and adenovirus were used to manipulate Nr1h4 expression. RNA-sequencing (RNA-seq), quantitative real-time PCR, enzyme-linked immunosorbent assay, Chromatin immunoprecipitation and western blotting were performed to further study the underlying mechanisms. We identified that NR1H4 was down-regulated during PD progression. In vitro experiments suggested that Nr1h4 knockdown led to inflammatory response, reactive oxygen species generation and astrocytes activation whereasNr1h4 overexpressionhad the opposite effects. The results of RNA-seq on astrocytes revealed that NR1H4 manipulated neuroinflammation in a CEBPβ/NF-κB dependent manner. Additionally, pharmacological activation of NR1H4 via Obeticholic acid ameliorated neuroinflammation and promoted neuronal survival. Our study first proved the neuroprotective effects of NR1H4against PD via inhibiting astrocyte activation and neuroinflammation in a CEBPβ/NF-κB dependent manner.
Collapse
Affiliation(s)
- Jingwen Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Hanshu Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Xinyu Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Shurui Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Qinwei Yu
- Wuhan Red Cross Hospital, Wuhan, Hubei, China
| | | | - Long Liu
- Wuhan Red Cross Hospital, Wuhan, Hubei, China
| | - Danfang Yu
- Wuhan Red Cross Hospital, Wuhan, Hubei, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China.
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China.
| |
Collapse
|
2
|
Qi L, Liu S, Fang Q, Qian C, Peng C, Liu Y, Yang P, Wu P, Shan L, Cui Q, Hua Q, Yang S, Ye C, Yang W, Li P, Xu X. Ginsenoside Rg3 Restores Mitochondrial Cardiolipin Homeostasis via GRB2 to Prevent Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403058. [PMID: 39159293 PMCID: PMC11497058 DOI: 10.1002/advs.202403058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/20/2024] [Indexed: 08/21/2024]
Abstract
Regulating cardiolipin to maintain mitochondrial homeostasis is a promising strategy for addressing Parkinson's disease (PD). Through a comprehensive screening and validation process involving multiple models, ginsenoside Rg3 (Rg3) as a compound capable of enhancing cardiolipin levels is identified. This augmentation in cardiolipin levels fosters mitochondrial homeostasis by bolstering mitochondrial unfolded protein response, promoting mitophagy, and enhancing mitochondrial oxidative phosphorylation. Consequently, this cascade enhances the survival of tyrosine hydroxylase positive (TH+) dopaminergic neurons, leading to an amelioration in motor performance within PD mouse models. Using limited proteolysis-small-molecule mapping combined with molecular docking analysis, it has confirmed Growth Factor Receptor-Bound Protein 2 (GRB2) as a molecular target for Rg3. Furthermore, these investigations reveal that Rg3 facilitates the interaction between GRB2 and TRKA (Neurotrophic Tyrosine Kinase, Receptor, Type 1), thus promotes EVI1 (Ecotropic Virus Integration Site 1 Protein Homolog) phosphorylation by ERK, subsequently increases CRLS1 (Cardiolipin Synthase 1) gene expression and boosts cardiolipin synthesis. The absence of GRB2 or CRLS1 significantly attenuates the beneficial effects of Rg3 on PD symptoms. Finally, Tenofovir Disoproxil Fumarate (TDF) that also promotes the binding between GRB2 and TRKA is further identified. The identified compounds, Rg3 and TDF, exhibit promising potential for the prevention of PD by bolstering cardiolipin expression and reinstating mitochondrial homeostasis.
Collapse
Affiliation(s)
- Li‐Feng‐Rong Qi
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Shuai Liu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
- Department of PharmacyThe Fourth Affiliated HospitalCenter for Innovative Traditional Chinese Medicine Target and New Drug ResearchInternational Institutes of MedicineZhejiang University School of MedicineYiwuZhejiang322000China
| | - Qiuyuan Fang
- Department of Biophysics and Department of Neurosurgery of the First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Cheng Qian
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Chao Peng
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai Advanced Research InstituteChinese Academy of ScienceShanghai201210China
- Shanghai Science Research CenterChinese Academy of SciencesShanghai201204China
| | - Yuci Liu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Peng Yang
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Ping Wu
- National Facility for Protein Science in ShanghaiZhangjiang LabShanghai Advanced Research InstituteChinese Academy of ScienceShanghai201210China
- Shanghai Science Research CenterChinese Academy of SciencesShanghai201204China
| | - Ling Shan
- Dept. Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesMeibergdreef 47Amsterdam1105BAthe Netherlands
| | - Qinghua Cui
- Department of Biomedical InformaticsSchool of Basic Medical SciencesKey Laboratory of Molecular Cardiovascular Sciences of the Ministry of EducationCenter for Non‐Coding RNA MedicinePeking University Health Science Center BeijingBeijing100191China
| | - Qian Hua
- School of Life SciencesBeijing University of Chinese MedicineBeijing100029China
| | - Sen Yang
- Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Cunqi Ye
- Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Wei Yang
- Department of PharmacyThe Fourth Affiliated HospitalCenter for Innovative Traditional Chinese Medicine Target and New Drug ResearchInternational Institutes of MedicineZhejiang University School of MedicineYiwuZhejiang322000China
| | - Ping Li
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Xiaojun Xu
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingJiangsu210009China
- Department of PharmacyThe Fourth Affiliated HospitalCenter for Innovative Traditional Chinese Medicine Target and New Drug ResearchInternational Institutes of MedicineZhejiang University School of MedicineYiwuZhejiang322000China
| |
Collapse
|
3
|
Li Z, Zhang Y, Ji M, Wu C, Zhang Y, Ji S. Targeting ferroptosis in neuroimmune and neurodegenerative disorders for the development of novel therapeutics. Biomed Pharmacother 2024; 176:116777. [PMID: 38795640 DOI: 10.1016/j.biopha.2024.116777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
Neuroimmune and neurodegenerative ailments impose a substantial societal burden. Neuroimmune disorders involve the intricate regulatory interactions between the immune system and the central nervous system. Prominent examples of neuroimmune disorders encompass multiple sclerosis and neuromyelitis optica. Neurodegenerative diseases result from neuronal degeneration or demyelination in the brain or spinal cord, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. The precise underlying pathogenesis of these conditions remains incompletely understood. Ferroptosis, a programmed form of cell death characterised by lipid peroxidation and iron overload, plays a pivotal role in neuroimmune and neurodegenerative diseases. In this review, we provide a detailed overview of ferroptosis, its mechanisms, pathways, and regulation during the progression of neuroimmune and neurodegenerative diseases. Furthermore, we summarise the impact of ferroptosis on neuroimmune-related cells (T cells, B cells, neutrophils, and macrophages) and neural cells (glial cells and neurons). Finally, we explore the potential therapeutic implications of ferroptosis inhibitors in diverse neuroimmune and neurodegenerative diseases.
Collapse
Affiliation(s)
- Zihao Li
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China
| | - Ye Zhang
- Department of Forensic Medicine, Shantou University Medical College (SUMC), Shantou, Guangdong, China
| | - Meiling Ji
- Department of Emergency, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 210002, China
| | - Chenglong Wu
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China
| | - Yanxing Zhang
- Department of Neurology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China.
| | - Senlin Ji
- Department of Neurology of Nanjing Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Translational Medicine Institute of Brain Disorders, Nanjing University, Nanjing, Jiangsu 210008, China.
| |
Collapse
|
4
|
Eberhard P, Kern M, Aichem M, Borlinghaus H, Klein K, Delp J, Suciu I, Moser B, Dietrich D, Leist M, Schreiber F. PathwayNexus: a tool for interactive metabolic data analysis. Bioinformatics 2024; 40:btae310. [PMID: 38724240 PMCID: PMC11153943 DOI: 10.1093/bioinformatics/btae310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/28/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
MOTIVATION High-throughput omics methods increasingly result in large datasets including metabolomics data, which are often difficult to analyse. RESULTS To help researchers to handle and analyse those datasets by mapping and investigating metabolomics data of multiple sampling conditions (e.g. different time points or treatments) in the context of pathways, PathwayNexus has been developed, which presents the mapping results in a matrix format, allowing users to easily observe the relations between the compounds and the pathways. It also offers functionalities like ranking, sorting, clustering, pathway views, and further analytical tools. Its primary objective is to condense large sets of pathways into smaller, more relevant subsets that align with the specific interests of the user. AVAILABILITY AND IMPLEMENTATION The methodology presented here is implemented in PathwayNexus, an open-source add-on for Vanted available at www.cls.uni-konstanz.de/software/pathway-nexus. CONTACT falk.schreiber@unikonstanz.de. SUPPLEMENTARY INFORMATION Website: www.cls.uni-konstanz.de/software/pathway-nexus.
Collapse
Affiliation(s)
- Philipp Eberhard
- Department of Computer and Information Science, University of Konstanz, Konstanz 78464, Germany
| | - Martin Kern
- Department of Computer and Information Science, University of Konstanz, Konstanz 78464, Germany
| | - Michael Aichem
- Department of Computer and Information Science, University of Konstanz, Konstanz 78464, Germany
| | - Hanna Borlinghaus
- Department of Computer and Information Science, University of Konstanz, Konstanz 78464, Germany
| | - Karsten Klein
- Department of Computer and Information Science, University of Konstanz, Konstanz 78464, Germany
| | - Johannes Delp
- Department of In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz 78464, Germany
| | - Ilinca Suciu
- Department of In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz 78464, Germany
| | - Benjamin Moser
- Department of Computer and Information Science, University of Konstanz, Konstanz 78464, Germany
| | - Daniel Dietrich
- Department of Human and Environmental Toxicology, University of Konstanz, Konstanz 78464, Germany
| | - Marcel Leist
- Department of In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz 78464, Germany
| | - Falk Schreiber
- Department of Computer and Information Science, University of Konstanz, Konstanz 78464, Germany
- Faculty of Information Technology, Monash University, Clayton, 3800, Australia
| |
Collapse
|
5
|
Sharma T, Kumar R, Mukherjee S. Neuronal Vulnerability to Degeneration in Parkinson's Disease and Therapeutic Approaches. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:715-730. [PMID: 37185323 DOI: 10.2174/1871527322666230426155432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 05/17/2023]
Abstract
Parkinson's disease is the second most common neurodegenerative disease affecting millions of people worldwide. Despite the crucial threat it poses, currently, no specific therapy exists that can completely reverse or halt the progression of the disease. Parkinson's disease pathology is driven by neurodegeneration caused by the intraneuronal accumulation of alpha-synuclein (α-syn) aggregates in Lewy bodies in the substantia nigra region of the brain. Parkinson's disease is a multiorgan disease affecting the central nervous system (CNS) as well as the autonomic nervous system. A bidirectional route of spreading α-syn from the gut to CNS through the vagus nerve and vice versa has also been reported. Despite our understanding of the molecular and pathophysiological aspects of Parkinson's disease, many questions remain unanswered regarding the selective vulnerability of neuronal populations, the neuromodulatory role of the locus coeruleus, and alpha-synuclein aggregation. This review article aims to describe the probable factors that contribute to selective neuronal vulnerability in Parkinson's disease, such as genetic predisposition, bioenergetics, and the physiology of neurons, as well as the interplay of environmental and exogenous modulators. This review also highlights various therapeutic strategies with cell transplants, through viral gene delivery, by targeting α-synuclein and aquaporin protein or epidermal growth factor receptors for the treatment of Parkinson's disease. The application of regenerative medicine and patient-specific personalized approaches have also been explored as promising strategies in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Tanushree Sharma
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Lucknow, Uttar Pradesh, India
- Molecular and Human Genetics, Banaras Hindu University Varanasi, Uttar Pradesh, India
| | - Rajnish Kumar
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Lucknow, Uttar Pradesh, India
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Sayali Mukherjee
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Lucknow, Uttar Pradesh, India
| |
Collapse
|
6
|
Suciu I, Delp J, Gutbier S, Suess J, Henschke L, Celardo I, Mayer TU, Amelio I, Leist M. Definition of the Neurotoxicity-Associated Metabolic Signature Triggered by Berberine and Other Respiratory Chain Inhibitors. Antioxidants (Basel) 2023; 13:49. [PMID: 38247474 PMCID: PMC10812665 DOI: 10.3390/antiox13010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
To characterize the hits from a phenotypic neurotoxicity screen, we obtained transcriptomics data for valinomycin, diethylstilbestrol, colchicine, rotenone, 1-methyl-4-phenylpyridinium (MPP), carbaryl and berberine (Ber). For all compounds, the concentration triggering neurite degeneration correlated with the onset of gene expression changes. The mechanistically diverse toxicants caused similar patterns of gene regulation: the responses were dominated by cell de-differentiation and a triggering of canonical stress response pathways driven by ATF4 and NRF2. To obtain more detailed and specific information on the modes-of-action, the effects on energy metabolism (respiration and glycolysis) were measured. Ber, rotenone and MPP inhibited the mitochondrial respiratory chain and they shared complex I as the target. This group of toxicants was further evaluated by metabolomics under experimental conditions that did not deplete ATP. Ber (204 changed metabolites) showed similar effects as MPP and rotenone. The overall metabolic situation was characterized by oxidative stress, an over-abundance of NADH (>1000% increase) and a re-routing of metabolism in order to dispose of the nitrogen resulting from increased amino acid turnover. This unique overall pattern led to the accumulation of metabolites known as biomarkers of neurodegeneration (saccharopine, aminoadipate and branched-chain ketoacids). These findings suggest that neurotoxicity of mitochondrial inhibitors may result from an ensemble of metabolic changes rather than from a simple ATP depletion. The combi-omics approach used here provided richer and more specific MoA data than the more common transcriptomics analysis alone. As Ber, a human drug and food supplement, mimicked closely the mode-of-action of known neurotoxicants, its potential hazard requires further investigation.
Collapse
Affiliation(s)
- Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Graduate School of Chemical Biology, University of Konstanz, 78464 Konstanz, Germany
| | - Johannes Delp
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Simon Gutbier
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Julian Suess
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Lars Henschke
- Graduate School of Chemical Biology, University of Konstanz, 78464 Konstanz, Germany
- Department of Molecular Genetics, University of Konstanz, 78464 Konstanz, Germany
| | - Ivana Celardo
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Thomas U. Mayer
- Department of Molecular Genetics, University of Konstanz, 78464 Konstanz, Germany
| | - Ivano Amelio
- Division for Systems Toxicology, Department of Biology, University of Konstanz, 78464 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| |
Collapse
|
7
|
Fidelis EM, Savall ASP, Mello JD, Quines CB, Comis-Neto AA, Sampaio TB, Denardin CC, de Ávila DS, Rosa SG, Pinton S. Purple pitanga extract (Eugenia uniflora) attenuates oxidative stress induced by MPTP. Metab Brain Dis 2023; 38:2615-2625. [PMID: 37921949 DOI: 10.1007/s11011-023-01318-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/25/2023] [Indexed: 11/05/2023]
Abstract
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) has been widely used due to its specific and reproducible neurotoxic effect on the nigrostriatal system, being considered a convenient model of dopaminergic neurodegeneration to study interventions therapeutics. The purple pitanga (Eugenia uniflora) is a polyphenol-rich fruit with antioxidant and antidepressant properties, among others. Therefore, this study investigated the effect of purple pitanga extract (PPE) on acute early oxidative stress induced by intranasal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration in rats. Male Wistar rats were pre-treated orally with PPE (1000 mg/kg) or vehicle. After 24 h, MPTP (0.1 mg/10µL/nostril) or vehicle was administered bilaterally into the animal's nostrils, and 6 h later, the olfactory bulb (OB), striatum (ST), and substantia nigra (SN) were collected to evaluate the oxidative stress parameters. Our findings revealed that OB and SN were the most affected areas after 6 h of MPTP infusion; an early increase in reactive oxygen species (ROS) levels was observed, while pretreatment with a single dose of PPE prevented this increment. No differences in thiobarbituric acid reactive species (TBARS) and 3-nitrotyrosine (3-NT) formation were observed, although 4-hydroxy-2-nonenal (4-HNE) levels increased, which is the most toxic form of lipid peroxidation, in the MPTP group. The PPE pretreatment could prevent this increase by increasing the NPSH levels previously decreased by MPTP. Furthermore, PPE prevents the Na+/K + ATPase strongly inhibited by MPTP, showing the neuroprotective capacity of the PPE by inhibiting the MPTP-generated oxidation. Thus, we demonstrated for the first time the antioxidant and neuroprotective effects of PPE against the early MPTP neurotoxicity.
Collapse
Affiliation(s)
| | - Anne Suely P Savall
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana, CEP 97500-970, RS, Brazil
| | - Jhuly Dornelles Mello
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana, CEP 97500-970, RS, Brazil
| | - Caroline Brandão Quines
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana, CEP 97500-970, RS, Brazil
- Regional University of the Northwest of the State of Rio Grande do Sul - Campus Ijuí, Ijuí, CEP 98700-000, RS, Brazil
| | | | | | | | - Daiana Silva de Ávila
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana, CEP 97500-970, RS, Brazil
| | - Suzan Gonçalves Rosa
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana, CEP 97500-970, RS, Brazil
| | - Simone Pinton
- Federal University of Pampa - Campus Uruguaiana, Uruguaiana, CEP 97500-970, RS, Brazil.
- Universidade Federal do Pampa - Campus Uruguaiana, Uruguaiana, CEP 97500-970, RS, Brazil.
| |
Collapse
|
8
|
Li M, Jiang H, Wang Y, Xu Z, Xu H, Chen Y, Zhu J, Lin Z, Zhang M. Effect of arctigenin on neurological diseases: A review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 315:116642. [PMID: 37236381 DOI: 10.1016/j.jep.2023.116642] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/25/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Arctium lappa L. is a common specie of Asteraceae. Its main active ingredient, Arctigenin (AG), in mature seeds exerts pharmacological effects on the Central Nervous System (CNS). AIM OF THE STUDY To review studies on the specific effects of the AG mechanism on various CNS diseases and elucidate signal transduction mechanisms and their pharmacological actions. MATERIALS AND METHODS This investigation reviewed the essential role of AG in treating neurological disorders. Basic information on Arctium lappa L. was retrieved from the Pharmacopoeia of the People's Republic of China. The related articles from 1981 to 2022 on the network database (including CNKI, PubMed, and Wan Fang and so on) were reviewed using AG and CNS diseases-related terms such as Arctigenin and Epilepsy. RESULTS It was confirmed that AG has a therapeutic effect on Alzheimer's disease, Glioma, infectious CNS diseases (such as Toxoplasma and Japanese Encephalitis Virus), Parkinson's disease, Epilepsy, etc. In these diseases, related experiments such as a Western blot analysis revealed that AG could alter the content of some key factors (such as the reduction of Aβ in Alzheimer's disease). However, in-vivo AG's metabolic process and possible metabolites are still undetermined. CONCLUSION Based on this review, the existing pharmacological research has indeed made objective progress to elucidate how AG prevents and treats CNS diseases, especially senile degenerative disease such as Alzheimer's diseases. It was revealed that AG could be used as a potential nervous system drug as it has a wide range of effects in theory with markedly high application value, especially in the elder group. However, the existing studies are limited to in-vitro experiments; therefore, little is known about how AG metabolizes and functions in-vivo, limiting its clinical application and requiring further research.
Collapse
Affiliation(s)
- Mopu Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haibin Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanan Wang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zidi Xu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hang Xu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuetong Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianghu Zhu
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China.
| | - Zhenlang Lin
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China.
| | - Min Zhang
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang, China.
| |
Collapse
|
9
|
Ückert AK, Rütschlin S, Gutbier S, Wörz NC, Miah MR, Martins AC, Hauer I, Holzer AK, Meyburg B, Mix AK, Hauck C, Aschner M, Böttcher T, Leist M. Identification of the bacterial metabolite aerugine as potential trigger of human dopaminergic neurodegeneration. ENVIRONMENT INTERNATIONAL 2023; 180:108229. [PMID: 37797477 PMCID: PMC10666548 DOI: 10.1016/j.envint.2023.108229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/14/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
The causes of nigrostriatal cell death in idiopathic Parkinson's disease are unknown, but exposure to toxic chemicals may play some role. We followed up here on suggestions that bacterial secondary metabolites might be selectively cytotoxic to dopaminergic neurons. Extracts from Streptomyces venezuelae were found to kill human dopaminergic neurons (LUHMES cells). Utilizing this model system as a bioassay, we identified a bacterial metabolite known as aerugine (C10H11NO2S; 2-[4-(hydroxymethyl)-4,5-dihydro-1,3-thiazol-2-yl]phenol) and confirmed this finding by chemical re-synthesis. This 2-hydroxyphenyl-thiazoline compound was previously shown to be a product of a wide-spread biosynthetic cluster also found in the human microbiome and in several pathogens. Aerugine triggered half-maximal dopaminergic neurotoxicity at 3-4 µM. It was less toxic for other neurons (10-20 µM), and non-toxic (at <100 µM) for common human cell lines. Neurotoxicity was completely prevented by several iron chelators, by distinct anti-oxidants and by a caspase inhibitor. In the Caenorhabditis elegans model organism, general survival was not affected by aerugine concentrations up to 100 µM. When transgenic worms, expressing green fluorescent protein only in their dopamine neurons, were exposed to aerugine, specific neurodegeneration was observed. The toxicant also exerted functional dopaminergic toxicity in nematodes as determined by the "basal slowing response" assay. Thus, our research has unveiled a bacterial metabolite with a remarkably selective toxicity toward human dopaminergic neurons in vitro and for the dopaminergic nervous system of Caenorhabditis elegans in vivo. These findings suggest that microbe-derived environmental chemicals should be further investigated for their role in the pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- Anna-Katharina Ückert
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Sina Rütschlin
- Department of Chemistry, Konstanz Research School Chemical Biology, Zukunftskolleg, University of Konstanz, 78457 Konstanz, Germany
| | - Simon Gutbier
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Nathalie Christine Wörz
- Faculty of Chemistry, Institute for Biological Chemistry & Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystems Science, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), 1090 Vienna, Austria; Doctoral School in Chemistry (DoSChem), University of Vienna, 1090 Vienna, Austria
| | - Mahfuzur R Miah
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 10641 Bronx, NY, United States
| | - Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 10641 Bronx, NY, United States; Department of Neuroscience, Albert Einstein College of Medicine, 10641 Bronx, NY, United States
| | - Isa Hauer
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Anna-Katharina Holzer
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Birthe Meyburg
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Ann-Kathrin Mix
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457 Konstanz, Germany
| | - Christof Hauck
- Lehrstuhl Zellbiologie, Universität Konstanz, Universitätsstraße 10, Postablage 621, 78457 Konstanz, Germany
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 10641 Bronx, NY, United States; Department of Neuroscience, Albert Einstein College of Medicine, 10641 Bronx, NY, United States
| | - Thomas Böttcher
- Department of Chemistry, Konstanz Research School Chemical Biology, Zukunftskolleg, University of Konstanz, 78457 Konstanz, Germany; Faculty of Chemistry, Institute for Biological Chemistry & Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystems Science, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), 1090 Vienna, Austria.
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
10
|
Barroso-Chinea P, Salas-Hernández J, Cruz-Muros I, López-Fernández J, Freire R, Afonso-Oramas D. Expression of RAD9B in the mesostriatal system of rats and humans: Overexpression in a 6-OHDA rat model of Parkinson's disease. Ann Anat 2023; 250:152135. [PMID: 37460044 DOI: 10.1016/j.aanat.2023.152135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/31/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder that affects primarily the dopaminergic (DAergic) neurons of the mesostriatal system, among other nuclei of the brain. Although it is considered an idiopathic disease, oxidative stress is believed to be involved in DAergic neuron death and therefore plays an important role in the onset and development of the disease. RAD9B is a paralog of the RAD9 checkpoint, sharing some similar functions related to DNA damage resistance and apoptosis, as well as the ability to form 9-1-1 heterotrimers with RAD1 and HUS1. METHODS In addition to immunohistochemistry, immunofluorescence and Western-blot analysis, we implemented Quantitative RT-PCR and in situ hybridization techniques. RESULTS We demonstrated RAD9B expression in rat and human mesencephalic DAergic cells using specific markers. Additionally, we observed significant overexpression of RAD9B mRNA (p<0.01) and protein (p<0.01) in the midbrain 48 h after inducing damage with 150 µg of 6-hydroxydopamine (6-OHDA) injected in a rat model of PD. Regarding protein expression, the increased levels were observed in neurons of the mesostriatal system and returned to normal 5 days post-injury. CONCLUSIONS This response to a neurotoxin, known to produce oxidative stress specifically on DAergic neurons indicates the potential importance of RAD9B in this highly vulnerable population to cell death. In this model, RAD9B function appears to provide neuroprotection, as the induced lesion resulted in only mild degeneration. This observation highlights the potential of RAD9B checkpoint protein as a valuable target for future therapeutic interventions aimed at promoting neuroprotection.
Collapse
Affiliation(s)
- Pedro Barroso-Chinea
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain; Instituto Universitario de Neurociencias (IUNE). Universidad de La Laguna, Tenerife, Spain.
| | - Josmar Salas-Hernández
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Ignacio Cruz-Muros
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Jonathan López-Fernández
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Raimundo Freire
- Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain; Fundación Canaria del Instituto de Investigación Sanitaria de Canarias (FIISC), Unidad de Investigación, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain; Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Domingo Afonso-Oramas
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain; Instituto Universitario de Neurociencias (IUNE). Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
11
|
Domenicale C, Magnabosco S, Morari M. Modeling Parkinson's disease in LRRK2 rodents. Neuronal Signal 2023; 7:NS20220040. [PMID: 37601008 PMCID: PMC10432857 DOI: 10.1042/ns20220040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 08/22/2023] Open
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are associated with familial and sporadic forms of Parkinson's disease (PD). Sporadic PD and LRRK2 PD share main clinical and neuropathological features, namely hypokinesia, degeneration of nigro-striatal dopamine neurons and α-synuclein aggregates in the form of Lewy bodies. Animals harboring the most common LRRK2 mutations, i.e. p.G2019S and p.R1441C/G, have been generated to replicate the parkinsonian phenotype and investigate the underlying pathogenic mechanisms. Disappointingly, however, LRRK2 rodents did not consistently phenocopy hypokinesia and nigro-striatal degeneration, or showed Lewy body-like aggregates. Instead, LRRK2 rodents manifested non-motor signs and dysregulated transmission at dopaminergic and non-dopaminergic synapses that are reminiscent of behavioral and functional network changes observed in the prodromal phase of the disease. LRRK2 rodents also manifested greater susceptibility to different parkinsonian toxins or stressors when subjected to dual-hit or multiple-hit protocols, confirming LRRK2 mutations as genetic risk factors. In conclusion, LRRK2 rodents represent a unique tool to identify the molecular mechanisms through which LRRK2 modulates the course and clinical presentations of PD and to study the interplay between genetic, intrinsic and environmental protective/risk factors in PD pathogenesis.
Collapse
Affiliation(s)
- Chiara Domenicale
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Stefano Magnabosco
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Michele Morari
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
12
|
Suciu I, Pamies D, Peruzzo R, Wirtz PH, Smirnova L, Pallocca G, Hauck C, Cronin MTD, Hengstler JG, Brunner T, Hartung T, Amelio I, Leist M. G × E interactions as a basis for toxicological uncertainty. Arch Toxicol 2023; 97:2035-2049. [PMID: 37258688 PMCID: PMC10256652 DOI: 10.1007/s00204-023-03500-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/17/2023] [Indexed: 06/02/2023]
Abstract
To transfer toxicological findings from model systems, e.g. animals, to humans, standardized safety factors are applied to account for intra-species and inter-species variabilities. An alternative approach would be to measure and model the actual compound-specific uncertainties. This biological concept assumes that all observed toxicities depend not only on the exposure situation (environment = E), but also on the genetic (G) background of the model (G × E). As a quantitative discipline, toxicology needs to move beyond merely qualitative G × E concepts. Research programs are required that determine the major biological variabilities affecting toxicity and categorize their relative weights and contributions. In a complementary approach, detailed case studies need to explore the role of genetic backgrounds in the adverse effects of defined chemicals. In addition, current understanding of the selection and propagation of adverse outcome pathways (AOP) in different biological environments is very limited. To improve understanding, a particular focus is required on modulatory and counter-regulatory steps. For quantitative approaches to address uncertainties, the concept of "genetic" influence needs a more precise definition. What is usually meant by this term in the context of G × E are the protein functions encoded by the genes. Besides the gene sequence, the regulation of the gene expression and function should also be accounted for. The widened concept of past and present "gene expression" influences is summarized here as Ge. Also, the concept of "environment" needs some re-consideration in situations where exposure timing (Et) is pivotal: prolonged or repeated exposure to the insult (chemical, physical, life style) affects Ge. This implies that it changes the model system. The interaction of Ge with Et might be denoted as Ge × Et. We provide here general explanations and specific examples for this concept and show how it could be applied in the context of New Approach Methodologies (NAM).
Collapse
Affiliation(s)
- Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated By the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - David Pamies
- Department of Biological Sciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Roberta Peruzzo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Petra H Wirtz
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, 78457, Constance, Germany
- Biological Work and Health Psychology, Department of Psychology, University of Konstanz, 78457, Constance, Germany
| | - Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | | | - Christof Hauck
- Department of Cell Biology, University of Konstanz, 78457, Constance, Germany
| | - Mark T D Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, 44139, Dortmund, Germany
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, 78457, Constance, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- CAAT Europe, University of Konstanz, 78457, Constance, Germany
| | - Ivano Amelio
- Division for Systems Toxicology, Department of Biology, University of Konstanz, 78457, Constance, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated By the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany.
- CAAT Europe, University of Konstanz, 78457, Constance, Germany.
| |
Collapse
|
13
|
Mirzac D, Kreis SL, Luhmann HJ, Gonzalez-Escamilla G, Groppa S. Translating Pathological Brain Activity Primers in Parkinson's Disease Research. RESEARCH (WASHINGTON, D.C.) 2023; 6:0183. [PMID: 37383218 PMCID: PMC10298229 DOI: 10.34133/research.0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
Translational experimental approaches that help us better trace Parkinson's disease (PD) pathophysiological mechanisms leading to new therapeutic targets are urgently needed. In this article, we review recent experimental and clinical studies addressing abnormal neuronal activity and pathological network oscillations, as well as their underlying mechanisms and modulation. Our aim is to enhance our knowledge about the progression of Parkinson's disease pathology and the timing of its symptom's manifestation. Here, we present mechanistic insights relevant for the generation of aberrant oscillatory activity within the cortico-basal ganglia circuits. We summarize recent achievements extrapolated from available PD animal models, discuss their advantages and limitations, debate on their differential applicability, and suggest approaches for transferring knowledge on disease pathology into future research and clinical applications.
Collapse
Affiliation(s)
- Daniela Mirzac
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Svenja L. Kreis
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Heiko J. Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Gabriel Gonzalez-Escamilla
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Sergiu Groppa
- Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| |
Collapse
|
14
|
Xing M, Li G, Liu Y, Yang L, Zhang Y, Zhang Y, Ding J, Lu M, Yu G, Hu G. Fucoidan from Fucus vesiculosus prevents the loss of dopaminergic neurons by alleviating mitochondrial dysfunction through targeting ATP5F1a. Carbohydr Polym 2023; 303:120470. [PMID: 36657849 DOI: 10.1016/j.carbpol.2022.120470] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Parkinson's disease is a neurodegenerative disease that is characterized by the loss of dopaminergic neurons. Fucoidan, which has emerged as a neuroprotective agent, is a marine-origin sulfated polysaccharide enriched in brown algae and sea cucumbers. However, variations in structural characteristics exist among fucoidans derived from different sources, resulting in a wide spectrum of biological effects. It is urgent to find the fucoidan with the strongest neuroprotective effect, and the mechanism needs to be further explored. We isolated and purified four different fucoidan species with different chemical structures and found that Type II fucoidan from Fucus vesiculosus (FvF) significantly improved mitochondrial dysfunction, prevented neuronal apoptosis, reduced dopaminergic neuron loss, and improved motor deficits in an 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model. Further mechanistic investigation revealed that the ATP5F1a protein is a key target responsible for alleviating mitochondrial dysfunction of FvF to exert neuroprotective effects. This study highlights the favorable properties of FvF for neuroprotection, making FvF a promising candidate for the treatment of PD.
Collapse
Affiliation(s)
- Meimei Xing
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Guoyun Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
| | - Yang Liu
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Luyao Yang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Youjiao Zhang
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yuruo Zhang
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211116, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211116, China.
| | - Guangli Yu
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China.
| | - Gang Hu
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211116, China.
| |
Collapse
|
15
|
Suciu I, Delp J, Gutbier S, Ückert AK, Spreng AS, Eberhard P, Karreman C, Schreiber F, Madjar K, Rahnenführer J, Celardo I, Amelio I, Leist M. Dynamic Metabolic and Transcriptional Responses of Proteasome-Inhibited Neurons. Antioxidants (Basel) 2023; 12:164. [PMID: 36671027 PMCID: PMC9854434 DOI: 10.3390/antiox12010164] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
Proteasome inhibition is associated with parkinsonian pathology in vivo and degeneration of dopaminergic neurons in vitro. We explored here the metabolome (386 metabolites) and transcriptome (3257 transcripts) regulations of human LUHMES neurons, following exposure to MG-132 [100 nM]. This proteasome inhibitor killed cells within 24 h but did not reduce viability for 12 h. Overall, 206 metabolites were changed in live neurons. The early (3 h) metabolome changes suggested a compromised energy metabolism. For instance, AMP, NADH and lactate were up-regulated, while glycolytic and citric acid cycle intermediates were down-regulated. At later time points, glutathione-related metabolites were up-regulated, most likely by an early oxidative stress response and activation of NRF2/ATF4 target genes. The transcriptome pattern confirmed proteostatic stress (fast up-regulation of proteasome subunits) and also suggested the progressive activation of additional stress response pathways. The early ones (e.g., HIF-1, NF-kB, HSF-1) can be considered a cytoprotective cellular counter-regulation, which maintained cell viability. For instance, a very strong up-regulation of AIFM2 (=FSP1) may have prevented fast ferroptotic death. For most of the initial period, a definite life-death decision was not taken, as neurons could be rescued for at least 10 h after the start of proteasome inhibition. Late responses involved p53 activation and catabolic processes such as a loss of pyrimidine synthesis intermediates. We interpret this as a phase of co-occurrence of protective and maladaptive cellular changes. Altogether, this combined metabolomics-transcriptomics analysis informs on responses triggered in neurons by proteasome dysfunction that may be targeted by novel therapeutic intervention in Parkinson's disease.
Collapse
Affiliation(s)
- Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
- Graduate School of Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Johannes Delp
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Simon Gutbier
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Anna-Katharina Ückert
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
- Graduate School of Biological Sciences, University of Konstanz, 78457 Konstanz, Germany
| | - Anna-Sophie Spreng
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
- Graduate School of Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Philipp Eberhard
- Department of Computer and Information Science, University of Konstanz, 78457 Konstanz, Germany
| | - Christiaan Karreman
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Falk Schreiber
- Department of Computer and Information Science, University of Konstanz, 78457 Konstanz, Germany
- Faculty of Information Technology, Monash University, Clayton 3800, Australia
| | - Katrin Madjar
- Department of Statistics, TU Dortmund University, 44221 Dortmund, Germany
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University, 44221 Dortmund, Germany
| | - Ivana Celardo
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
| | - Ivano Amelio
- Division for Systems Toxicology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany
- The Center for Alternatives to Animal Testing in Europe, University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
16
|
In Silico Evaluation and In Vitro Determination of Neuroprotective and MAO-B Inhibitory Effects of Pyrrole-Based Hydrazones: A Therapeutic Approach to Parkinson's Disease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238485. [PMID: 36500572 PMCID: PMC9737692 DOI: 10.3390/molecules27238485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/19/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022]
Abstract
Parkinson's disease is a huge burden in modern medicinal practice. A serious drawback of current antiparkinsonian therapy is its symptomatic nature. This directed our investigations in the search for new more potent derivatives, affecting not only the loss of dopaminergic neurons but also the oxidative damage of neuronal cells. Thus in vitro neurotoxicity and neuroprotective analysis on a group of N-pyrrolyl hydrazide-hydrazones were performed. The neurotoxicity of the target derivatives was determined on a subcellular level in isolated rat synaptosomes, mitochondria and microsomes determining their effect on cellular vitality, GSH depletion and MDA production. The neuroprotective effects of the evaluated hydrazones were measured in three models of induced oxidative stress: 6-OHDA, t-BuOOH and Fe2+/AA-induced lipid peroxidation. Molecular docking simulations along with in vitro evaluation of MAO-B inhibitory potential of the target molecules were also performed. The results identified the ethyl 5-(4-bromophenyl)-1-(3-hydrazinyl-3-oxopropyl)-2-methyl-1H-pyrrole-3-carboxylate (12) as the most promising compound with the lowest neurotoxicity and highest neuroprotection on all evaluated parameters and inhibiting the hMAOB enzyme by 50%, comparable with the activity of the reference, Selegiline. The compatibility of the in silico and in vitro evaluations is a good prerequisite for these methods to be applied in future assessment of pyrrole-based compounds as anti-Parkinson agents.
Collapse
|
17
|
Song J, Liu L, Li Z, Mao T, Zhang J, Zhou L, Chen X, Shang Y, Sun T, Luo Y, Jiang Y, Tan D, Tong X, Dai F. Lycium barbarum polysaccharide improves dopamine metabolism and symptoms in an MPTP-induced model of Parkinson's disease. BMC Med 2022; 20:412. [PMID: 36303171 PMCID: PMC9615188 DOI: 10.1186/s12916-022-02621-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/20/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disease in middle-aged and elderly populations, whereas there is no cure for PD so far. Novel animal models and medications await development to elucidate the aetiology of PD and attenuate the symptoms, respectively. METHODS A neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), was used in the current study to establish a PD pathologic model in silkworms. The time required to complete specific behaviours was recorded. Dopamine content was detected by ultra-performance liquid chromatography (UPLC). The activity of insect tyrosine hydroxylase (TH) was determined using a double-antibody sandwich method. Oxidative stress was assessed by changes in antioxidant enzyme activity and the content of oxidative products. RESULTS MPTP-treated silkworms were characterized by impaired motor ability, reduced dopamine content, and elevated oxidative stress level. The expression of TH, a dopamine biosynthetic enzyme within dopaminergic neurons in the brain, was significantly reduced, indicating that dopaminergic neurons were damaged. Moreover, MPTP-induced motility impairment and reduced dopamine level in the silkworm PD model could be rescued after feeding a combination of levodopa (L-dopa [LD]) and carbidopa (CD). MPTP-induced oxidative damage was also alleviated, in ways consistent with other PD animal models. Interestingly, administration of Lycium barbarum polysaccharide (LBP) improved the motor ability, dopamine level, and TH activity, and the oxidative damage was concomitantly reduced in the silkworm PD model. CONCLUSIONS This study provides a promising animal model for elucidating the pathogenesis of PD, as well as a relevant preliminary drug screening (e.g., LBP) and evaluation.
Collapse
Affiliation(s)
- Jiangbo Song
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Lian Liu
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Zhiquan Li
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Ting Mao
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Jianfei Zhang
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Lei Zhou
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Xin Chen
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Yunzhu Shang
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Tao Sun
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Yuxin Luo
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Yu Jiang
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Duan Tan
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Xiaoling Tong
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Fangyin Dai
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
18
|
Liu Y, Luo J, Liu Y, Liu W, Yu G, Huang Y, Yang Y, Chen X, Chen T. Brain-Targeted Biomimetic Nanodecoys with Neuroprotective Effects for Precise Therapy of Parkinson's Disease. ACS CENTRAL SCIENCE 2022; 8:1336-1349. [PMID: 36188350 PMCID: PMC9523773 DOI: 10.1021/acscentsci.2c00741] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Indexed: 06/16/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the gradual loss of dopaminergic neurons in the substantia nigra and the accumulation of α-synuclein aggregates called Lewy bodies. Here, nanodecoys were designed from a rabies virus polypeptide with a 29 amino acid (RVG29)-modified red blood cell membrane (RBCm) to encapsulate curcumin nanocrystals (Cur-NCs), which could effectively protect dopaminergic neurons. The RVG29-RBCm/Cur-NCs nanodecoys effectively escaped from reticuloendothelial system (RES) uptake, enabled prolonged blood circulation, and enhanced blood-brain barrier (BBB) crossing after systemic administration. Cur-NCs loaded inside the nanodecoys exhibited the recovery of dopamine levels, inhibition of α-synuclein aggregation, and reversal of mitochondrial dysfunction in PD mice. These findings indicate the promising potential of biomimetic nanodecoys in treating PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Yao Liu
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, Guangzhou 510405, China
| | - Jingshan Luo
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, Guangzhou 510405, China
| | - Yujing Liu
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, Guangzhou 510405, China
| | - Wen Liu
- State
Key Laboratory of Quality Research in Chinese Medicine, Institute
of Chinese Medical Sciences, University
of Macau, Macau 999078, China
| | - Guangtao Yu
- Stomatological
Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuting Huang
- Institute
of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Yang
- Institute
of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaojia Chen
- State
Key Laboratory of Quality Research in Chinese Medicine, Institute
of Chinese Medical Sciences, University
of Macau, Macau 999078, China
| | - Tongkai Chen
- Science
and Technology Innovation Center, Guangzhou
University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
19
|
Huang M, Bargues-Carot A, Riaz Z, Wickham H, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Impact of Environmental Risk Factors on Mitochondrial Dysfunction, Neuroinflammation, Protein Misfolding, and Oxidative Stress in the Etiopathogenesis of Parkinson's Disease. Int J Mol Sci 2022; 23:10808. [PMID: 36142718 PMCID: PMC9505762 DOI: 10.3390/ijms231810808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
As a prevalent progressive neurodegenerative disorder, Parkinson's disease (PD) is characterized by the neuropathological hallmark of the loss of nigrostriatal dopaminergic (DAergic) innervation and the appearance of Lewy bodies with aggregated α-synuclein. Although several familial forms of PD have been reported to be associated with several gene variants, most cases in nature are sporadic, triggered by a complex interplay of genetic and environmental risk factors. Numerous epidemiological studies during the past two decades have shown positive associations between PD and several environmental factors, including exposure to neurotoxic pesticides/herbicides and heavy metals as well as traumatic brain injury. Other environmental factors that have been implicated as potential risk factors for PD include industrial chemicals, wood pulp mills, farming, well-water consumption, and rural residence. In this review, we summarize the environmental toxicology of PD with the focus on the elaboration of chemical toxicity and the underlying pathogenic mechanisms associated with exposure to several neurotoxic chemicals, specifically 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), rotenone, paraquat (PQ), dichloro-diphenyl-trichloroethane (DDT), dieldrin, manganese (Mn), and vanadium (V). Our overview of the current findings from cellular, animal, and human studies of PD provides information for possible intervention strategies aimed at halting the initiation and exacerbation of environmentally linked PD.
Collapse
Affiliation(s)
- Minhong Huang
- Department of Biomedical Sciences, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Alejandra Bargues-Carot
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Zainab Riaz
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Hannah Wickham
- Department of Biomedical Sciences, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
| | - Gary Zenitsky
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Huajun Jin
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Vellareddy Anantharam
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Arthi Kanthasamy
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Anumantha G. Kanthasamy
- Department of Biomedical Sciences, Iowa State University, 2062 Veterinary Medicine Building, Ames, IA 50011, USA
- Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| |
Collapse
|
20
|
Cheng G, Liu X, Liu Y, Liu Y, Ma R, Luo J, Zhou X, Wu Z, Liu Z, Chen T, Yang Y. Ultrasmall Coordination Polymers for Alleviating ROS-Mediated Inflammatory and Realizing Neuroprotection against Parkinson's Disease. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9781323. [PMID: 35958109 PMCID: PMC9343083 DOI: 10.34133/2022/9781323] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/17/2022] [Indexed: 01/14/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease globally, and there is currently no effective treatment for this condition. Excessive accumulation of reactive oxygen species (ROS) and neuroinflammation are major contributors to PD pathogenesis. Herein, ultrasmall nanoscale coordination polymers (NCPs) coordinated by ferric ions and natural product curcumin (Cur) were exploited, showing efficient neuroprotection by scavenging excessive radicals and suppressing neuroinflammation. In a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse PD model, such ultrasmall Fe-Cur NCPs with prolonged blood circulation and BBB traversing capability could effectively alleviate oxidative stress, mitochondrial dysfunction, and inflammatory condition in the midbrain and striatum to reduce PD symptoms. Thus, this study puts forth a unique type of therapeutics-based NCPs that could be used for safe and efficient treatment of PD with potential in clinical translation.
Collapse
Affiliation(s)
- Guowang Cheng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xueliang Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yujing Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yao Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Rui Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jingshan Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xinyi Zhou
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
21
|
Marchetti B, Giachino C, Tirolo C, Serapide MF. "Reframing" dopamine signaling at the intersection of glial networks in the aged Parkinsonian brain as innate Nrf2/Wnt driver: Therapeutical implications. Aging Cell 2022; 21:e13575. [PMID: 35262262 PMCID: PMC9009237 DOI: 10.1111/acel.13575] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/11/2022] [Accepted: 02/06/2022] [Indexed: 11/30/2022] Open
Abstract
Dopamine (DA) signaling via G protein-coupled receptors is a multifunctional neurotransmitter and neuroendocrine-immune modulator. The DA nigrostriatal pathway, which controls the motor coordination, progressively degenerates in Parkinson's disease (PD), a most common neurodegenerative disorder (ND) characterized by a selective, age-dependent loss of substantia nigra pars compacta (SNpc) neurons, where DA itself is a primary source of oxidative stress and mitochondrial impairment, intersecting astrocyte and microglial inflammatory networks. Importantly, glia acts as a preferential neuroendocrine-immune DA target, in turn, counter-modulating inflammatory processes. With a major focus on DA intersection within the astrocyte-microglial inflammatory network in PD vulnerability, we herein first summarize the characteristics of DA signaling systems, the propensity of DA neurons to oxidative stress, and glial inflammatory triggers dictating the vulnerability to PD. Reciprocally, DA modulation of astrocytes and microglial reactivity, coupled to the synergic impact of gene-environment interactions, then constitute a further level of control regulating midbrain DA neuron (mDAn) survival/death. Not surprisingly, within this circuitry, DA converges to modulate nuclear factor erythroid 2-like 2 (Nrf2), the master regulator of cellular defense against oxidative stress and inflammation, and Wingless (Wnt)/β-catenin signaling, a key pathway for mDAn neurogenesis, neuroprotection, and immunomodulation, adding to the already complex "signaling puzzle," a novel actor in mDAn-glial regulatory machinery. Here, we propose an autoregulatory feedback system allowing DA to act as an endogenous Nrf2/Wnt innate modulator and trace the importance of DA receptor agonists applied to the clinic as immune modifiers.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC)Pharmacology SectionMedical SchoolUniversity of CataniaCataniaItaly
- OASI Research Institute‐IRCCS, Troina (EN), ItalyTroinaItaly
| | | | - Cataldo Tirolo
- OASI Research Institute‐IRCCS, Troina (EN), ItalyTroinaItaly
| | - Maria F. Serapide
- Department of Biomedical and Biotechnological Sciences (BIOMETEC)Pharmacology SectionMedical SchoolUniversity of CataniaCataniaItaly
| |
Collapse
|
22
|
Wu X, Ren Y, Wen Y, Lu S, Li H, Yu H, Li W, Zou F. Deacetylation of ZKSCAN3 by SIRT1 induces autophagy and protects SN4741 cells against MPP +-induced oxidative stress. Free Radic Biol Med 2022; 181:82-97. [PMID: 35124181 DOI: 10.1016/j.freeradbiomed.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/28/2021] [Accepted: 02/01/2022] [Indexed: 11/20/2022]
Abstract
Mitochondrial dysfunction, oxidative stress and misfolded protein aggregation are related to autophagy-lysosomal dysregulation and contribute to the pathogenesis of Parkinson' s disease (PD). ZKSCAN3, a transcriptional repressor, plays a crucial role in autophagy and lysosomal biogenesis. However, the role and modification of ZKSCAN3 in the defection of ALP, along with the molecular mechanism involved in pathogenesis of PD, still remain unclear. In this study, we demonstrated that cellular reactive oxygen species (ROS) generated by MPP+ exposure and the resulting oxidative damage were counteracted by SIRT1-ZKSCAN3 pathway induction. Here we showed that nuclear ZKSCAN3 significantly increased in ventral midbrain of MPTP-treated mice and MPP+-treated SN4741 cells. Knockdown of ZKSCAN3 alleviated MPP+-induced ALP defect, Tyrosine Hydroxylase (TH) declination and neuronal death. NAC, a ROS scavenger, reduced the nuclear translocation of ZKSCAN3 and sequentially improved ALP function in MPP+-treated SN4741 cells. SRT2104, a SIRT1 activator, attenuated impairment of ALP in MPP+-treated SN47417 cells through decreasing nuclear accumulation of ZKSCAN3 and protected dopaminergic neurons from MPTP injury. Moreover, SRT2104 relieved impairment in locomotor activities and coordination skills upon treatment of MPTP in C57/BL6J mice through behavior tests including rotarod, pole climbing and grid. Furthermore, ZKSCAN3 was a novel substrate of SIRT1 which was deacetylated at lysine 148 residues by SIRT1. This subsequently facilitated the shuttling of ZKSCAN3 to the cytoplasm. Therefore, our study identifies a novel acetylation-dependent regulatory mechanism of nuclear translocation of ZKSCAN3. It results in autophagy-lysosomal dysfunction and then leads to DA neuronal death in MPTP/MPP+ model of PD.
Collapse
Affiliation(s)
- Xian Wu
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Yixian Ren
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China; Evaluation and Monitoring Center of Occupational Health, Guangzhou Twelfth People's Hospital, Guangzhou, PR China.
| | - Yue Wen
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Sixin Lu
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Huihui Li
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Honglin Yu
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Wenjun Li
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
23
|
Cao X, Guo J, Mochizuki H, Xu D, Zhang T, Han H, Ma T, Qi M, He J. Circular RNA circ_0070441 regulates MPP +-triggered neurotoxic effect in SH-SY5Y cells via miR-626/IRS2 axis. Metab Brain Dis 2022; 37:513-524. [PMID: 34748128 DOI: 10.1007/s11011-021-00869-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/01/2021] [Indexed: 02/07/2023]
Abstract
Circular RNAs (circRNAs) was suggested to play crucial regulatory roles in various human diseases, including Parkinson's disease (PD). This research aimed to investigate the function and potential mechanism of circ_0070441 in PD. MPP+ (1-methyl-4-phenylpyridinium)-treated SH-SY5Y cells was used as an in vitro cellular PD model. The expressions of circ_0070441, microRNA (miR)-626 and insulin receptor substrate 2 (IRS2) were measured by quantitative real-time polymerase chain reaction (RT-qPCR) or western blot. Cell Counting Kit-8 (CCK-8) assay, Cytotoxicity Detection Kit (Lactate Dehydrogenase), flow cytometry and Caspase-3 Assay Kit were used to detect cell viability, LDH release, cell apoptosis and caspase-3 activity, respectively. The levels of inflammation-related factors were detected by enzyme-linked immunosorbent assay (ELISA). The correlation among circ_0070441, miR-626 and IRS2 were confirmed by dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull-down assay. The levels of circ_0070441 and IRS2 were increased while miR-626 expression was decreased in MPP+-treated SH-SY5Y cells in dose- and time-dependent manners. Depletion of circ_0070441 alleviated MPP+-triggered neuronal damage by regulating cell apoptosis and inflammation. Circ_0070441 acted as a sponge for miR-626, and IRS2 was a target of miR-626. Besides, the neuroprotective effects of circ_0070441 knockdown or miR-626 overexpression were partly overturned by the suppression of miR-626 or IRS2 overexpression. Moreover, circ_0070441 upregulated IRS2 expression by interacting with miR-626. In summary, circ_0070441 aggravated MPP+-triggered neurotoxic effect in SH-SY5Y cells by regulating miR-626/IRS2 axis.
Collapse
Affiliation(s)
- Xuqing Cao
- Department of Neurology, People's Hospital of Ningxia Hui Autonomous Region, Zhengyuan North Street, Jinfeng District, Yinchuan, 750002, China.
| | - Jiangtao Guo
- Department of Rheumatogy and Immunology, People's Hospital of Ningxia Hui Autonomous Region, 750002, Yinchuan, China
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, 550-0004, Osaka, Japan
| | - Dong Xu
- Department of Neurology, People's Hospital of Ningxia Hui Autonomous Region, Zhengyuan North Street, Jinfeng District, Yinchuan, 750002, China
| | - Tao Zhang
- Department of Neurology, People's Hospital of Ningxia Hui Autonomous Region, Zhengyuan North Street, Jinfeng District, Yinchuan, 750002, China
| | - Haiping Han
- Department of Neurology, People's Hospital of Ningxia Hui Autonomous Region, Zhengyuan North Street, Jinfeng District, Yinchuan, 750002, China
| | - Tingjie Ma
- Department of Neurology, People's Hospital of Ningxia Hui Autonomous Region, Zhengyuan North Street, Jinfeng District, Yinchuan, 750002, China
| | - Mingshan Qi
- Department of Neurology, People's Hospital of Ningxia Hui Autonomous Region, Zhengyuan North Street, Jinfeng District, Yinchuan, 750002, China
| | - Jing He
- Department of Neurology, People's Hospital of Ningxia Hui Autonomous Region, Zhengyuan North Street, Jinfeng District, Yinchuan, 750002, China
| |
Collapse
|
24
|
Ostapiuk A, Urbanska EM. Kynurenic acid in neurodegenerative disorders-unique neuroprotection or double-edged sword? CNS Neurosci Ther 2022; 28:19-35. [PMID: 34862742 PMCID: PMC8673711 DOI: 10.1111/cns.13768] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/24/2022] Open
Abstract
AIMS The family of kynurenine pathway (KP) metabolites includes compounds produced along two arms of the path and acting in clearly opposite ways. The equilibrium between neurotoxic kynurenines, such as 3-hydroxykynurenine (3-HK) or quinolinic acid (QUIN), and neuroprotective kynurenic acid (KYNA) profoundly impacts the function and survival of neurons. This comprehensive review summarizes accumulated evidence on the role of KYNA in Alzheimer's, Parkinson's and Huntington's diseases, and discusses future directions of potential pharmacological manipulations aimed to modulate brain KYNA. DISCUSSION The synthesis of specific KP metabolites is tightly regulated and may considerably vary under physiological and pathological conditions. Experimental data consistently imply that shift of the KP to neurotoxic branch producing 3-HK and QUIN formation, with a relative or absolute deficiency of KYNA, is an important factor contributing to neurodegeneration. Targeting specific brain regions to maintain adequate KYNA levels seems vital; however, it requires the development of precise pharmacological tools, allowing to avoid the potential cognitive adverse effects. CONCLUSIONS Boosting KYNA levels, through interference with the KP enzymes or through application of prodrugs/analogs with high bioavailability and potency, is a promising clinical approach. The use of KYNA, alone or in combination with other compounds precisely influencing specific populations of neurons, is awaiting to become a significant therapy for neurodegenerative disorders.
Collapse
Affiliation(s)
- Aleksandra Ostapiuk
- Laboratory of Cellular and Molecular PharmacologyDepartment of Experimental and Clinical PharmacologyMedical University of LublinLublinPoland
- Present address:
Department of Clinical Digestive OncologyKU LeuvenLeuvenBelgium
| | - Ewa M. Urbanska
- Laboratory of Cellular and Molecular PharmacologyDepartment of Experimental and Clinical PharmacologyMedical University of LublinLublinPoland
| |
Collapse
|
25
|
Novello S, Mercatelli D, Albanese F, Domenicale C, Brugnoli A, D'Aversa E, Vantaggiato S, Dovero S, Murtaj V, Presotto L, Borgatti M, Shimshek DR, Bezard E, Moresco RM, Belloli S, Morari M. In vivo susceptibility to energy failure parkinsonism and LRRK2 kinase activity. Neurobiol Dis 2021; 162:105579. [PMID: 34871735 DOI: 10.1016/j.nbd.2021.105579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/08/2021] [Accepted: 12/02/2021] [Indexed: 12/31/2022] Open
Abstract
The G2019S mutation of LRRK2 represents a risk factor for idiopathic Parkinson's disease. Here, we investigate whether LRRK2 kinase activity regulates susceptibility to the environmental toxin 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine (MPTP). G2019S knock-in mice (bearing enhanced kinase activity) showed greater nigro-striatal degeneration compared to LRRK2 knock-out, LRRK2 kinase-dead and wild-type mice following subacute MPTP treatment. LRRK2 kinase inhibitors PF-06447475 and MLi-2, tested under preventive or therapeutic treatments, protected against nigral dopamine cell loss in G2019S knock-in mice. MLi-2 also rescued striatal dopaminergic terminal degeneration in both G2019S knock-in and wild-type mice. Immunoblot analysis of LRRK2 Serine935 phosphorylation levels confirmed target engagement of LRRK2 inhibitors. However, MLi-2 abolished phosphoSerine935 levels in the striatum and midbrain of both wild-type and G2019S knock-in mice whereas PF-06447475 partly reduced phosphoSerine935 levels in the midbrain of both genotypes. In vivo and ex vivo uptake of the 18-kDa translocator protein (TSPO) ligand [18F]-VC701 revealed a similar TSPO binding in MPTP-treated wild-type and G2019S knock-in mice which was consistent with an increased GFAP striatal expression as revealed by Real Time PCR. We conclude that LRRK2 G2019S, likely through enhanced kinase activity, confers greater susceptibility to mitochondrial toxin-induced parkinsonism. LRRK2 kinase inhibitors are neuroprotective in this model.
Collapse
Affiliation(s)
- Salvatore Novello
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy.
| | - Daniela Mercatelli
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy; Technopole of Ferrara, LTTA Laboratory for Advanced Therapies, 44121 Ferrara, Italy.
| | - Federica Albanese
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy.
| | - Chiara Domenicale
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy.
| | - Alberto Brugnoli
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy.
| | - Elisabetta D'Aversa
- Department of Life Science and Biotechnology, University of Ferrara, 44121 Ferrara, Italy.
| | - Silvia Vantaggiato
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Sandra Dovero
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.
| | - Valentina Murtaj
- Nuclear Medicine Department, San Raffaele Scientific Institute, Milan, Italy; PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano Bicocca, Monza, Italy; Medicine and Surgery Department, University of Milano Bicocca, Monza, Italy.
| | - Luca Presotto
- Nuclear Medicine Department, San Raffaele Scientific Institute, Milan, Italy.
| | - Monica Borgatti
- Department of Life Science and Biotechnology, University of Ferrara, 44121 Ferrara, Italy.
| | - Derya R Shimshek
- Department of Neuroscience, Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4002 Basel, Switzerland.
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.
| | - Rosa Maria Moresco
- Nuclear Medicine Department, San Raffaele Scientific Institute, Milan, Italy; Medicine and Surgery Department, University of Milano Bicocca, Monza, Italy; Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.
| | - Sara Belloli
- Nuclear Medicine Department, San Raffaele Scientific Institute, Milan, Italy; Medicine and Surgery Department, University of Milano Bicocca, Monza, Italy; Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.
| | - Michele Morari
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
26
|
Vidyadhara DJ, Yarreiphang H, Raju TR, Alladi PA. Differences in Neuronal Numbers, Morphology, and Developmental Apoptosis in Mice Nigra Provide Experimental Evidence of Ontogenic Origin of Vulnerability to Parkinson's Disease. Neurotox Res 2021; 39:1892-1907. [PMID: 34762290 DOI: 10.1007/s12640-021-00439-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 10/19/2022]
Abstract
Parkinson disease (PD) prevalence varies by ethnicity. In an earlier study, we replicated the reduced vulnerability to PD in an admixed population, using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-susceptible C57BL/6 J, MPTP-resistant CD-1 and their F1 crossbreds. In the present study, we investigated if the differences have a developmental origin. Substantia nigra was evaluated at postnatal days 2 (P2), P6, P10, P14, P18, and P22. C57BL/6 J mice had smaller nigra and fewer dopaminergic neurons than the CD-1 and crossbreds at P2, which persisted through development. A significant increase in numbers and nigral volume was observed across strains until P14. A drastic decline thereafter was specific to C57BL/6 J. CD-1 and crossbreds retained their numbers from P14 to stabilize with supernumerary neurons at adulthood. The neuronal size increased gradually to attain adult morphology at P10 in the resistant strains, vis-à-vis at P22 in C57BL/6 J. Accordingly, in comparison to C57BL/6 J, the nigra of CD-1 and reciprocal crossbreds possessed cytomorphological features of resilience, since birth. The considerably lesser dopaminergic neuronal loss in the CD-1 and crossbreds was seen at P2 and P14 and thereafter was complemented by attenuated developmental cell death. The differences in programmed cell death were confirmed by reduced TUNEL labelling, AIF, and caspase-3 expression. GDNF expression aligned with the cell death pattern at P2 and P14 in both nigra and striatum. Earlier maturity of nigra and its neurons appears to be better features that reflect as MPTP resistance at adulthood. Thus, variable MPTP vulnerability in mice and also differential susceptibility to PD in humans may arise early during nigral development.
Collapse
Affiliation(s)
- D J Vidyadhara
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
- Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Haorei Yarreiphang
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Trichur R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Phalguni Anand Alladi
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India.
- Formerly at Department of Neurophysiology, National Institute of Mental Health and Neuro-Sciences, Hosur Road, Bangalore, India.
| |
Collapse
|
27
|
The Positive Role and Mechanism of Herbal Medicine in Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9923331. [PMID: 34567415 PMCID: PMC8457986 DOI: 10.1155/2021/9923331] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/23/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease, manifested by the progressive functional impairment of the midbrain nigral dopaminergic neurons. Due to the unclear underlying pathogenesis, disease-modifying drugs for PD remain elusive. In Asia, such as in China and India, herbal medicines have been used in the treatment of neurodegenerative disease for thousands of years, which recently attracted considerable attention because of the development of curative drugs for PD. In this review, we first summarized the pathogenic factors of PD including protein aggregation, mitochondrial dysfunction, ion accumulation, neuroinflammation, and oxidative stress, and the related recent advances. Secondly, we summarized 32 Chinese herbal medicines (belonging to 24 genera, such as Acanthopanax, Alpinia, and Astragalus), 22 Chinese traditional herbal formulations, and 3 Indian herbal medicines, of which the ethanol/water extraction or main bioactive compounds have been extensively investigated on PD models both in vitro and in vivo. We elaborately provided pictures of the representative herbs and the structural formula of the bioactive components (such as leutheroside B and astragaloside IV) of the herbal medicines. Also, we specified the potential targets of the bioactive compounds or extractions of herbs in view of the signaling pathways such as PI3K, NF-κB, and AMPK which are implicated in oxidative and inflammatory stress in neurons. We consider that this knowledge of herbal medicines or their bioactive components can be favorable for the development of disease-modifying drugs for PD.
Collapse
|
28
|
Liu Y, Hong H, Xue J, Luo J, Liu Q, Chen X, Pan Y, Zhou J, Liu Z, Chen T. Near-Infrared Radiation-Assisted Drug Delivery Nanoplatform to Realize Blood-Brain Barrier Crossing and Protection for Parkinsonian Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:37746-37760. [PMID: 34318658 DOI: 10.1021/acsami.1c12675] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Mitochondrial dysfunction, which is directly involved in Parkinson's disease (PD), is characterized by the production of reactive oxygen species (ROS) and aberrant energy metabolism. Thus, regulating mitochondrial function might be an effective strategy to treat PD. However, the blood-brain barrier (BBB) presents a significant challenge for the intracerebral delivery of drugs. Here, we synthesized a zeolitic imidazolate framework 8-coated Prussian blue nanocomposite (ZIF-8@PB), which was encapsulated with quercetin (QCT), a natural antioxidant, to treat PD. ZIF-8@PB-QCT exhibited superior near-infrared radiation (NIR) response and penetrated through the BBB to the site of mitochondrial damage guided by the photothermal effect. In the mice model of PD, the QCT released from ZIF-8@PB-QCT significantly increased the adenosine triphosphate levels, reduced the oxidative stress levels, and reversed dopaminergic neuronal damage as well as PD-related behavioral deficits without any damage to the normal tissues. Furthermore, we explored the underlying neuroprotective mechanism of ZIF-8@PB-QCT that was mediated by activating the PI3K/Akt signaling pathway. Thus, combined with noninvasive NIR radiation, the biocompatible ZIF-8@PB-QCT nanocomposite could be used to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Yao Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Honghai Hong
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Jincheng Xue
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Jingshan Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qiao Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jingwei Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zeming Liu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
29
|
Dionísio PA, Amaral JD, Rodrigues CMP. Oxidative stress and regulated cell death in Parkinson's disease. Ageing Res Rev 2021; 67:101263. [PMID: 33540042 DOI: 10.1016/j.arr.2021.101263] [Citation(s) in RCA: 181] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. Motor deficits usually associated with PD correlate with dopaminergic axonal neurodegeneration starting at the striatum, which is then followed by dopaminergic neuronal death in the substantia nigra pars compacta (SN), with both events occurring already at the prodromal stage. We will overview the main physiological characteristics responsible for the higher susceptibility of the nigrostriatal circuit to mitochondrial dysfunction and oxidative stress, as hinted by the acting mechanisms of the PD-causing neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Then, we will present multiple lines of evidence linking several cell death mechanisms involving mitochondria and production of reactive oxygen species to neuronal loss in PD, namely intrinsic and extrinsic apoptosis, necroptosis, ferroptosis, parthanatos and mitochondrial permeability transition-driven necrosis. We will focus on gathered data from postmortem PD samples and relevant in vivo models, especially MPTP-based models.
Collapse
Affiliation(s)
- P A Dionísio
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, 1649-003, Portugal
| | - J D Amaral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, 1649-003, Portugal
| | - C M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, 1649-003, Portugal.
| |
Collapse
|
30
|
Dutta N, Ghosh S, Nelson VK, Sareng HR, Majumder C, Mandal SC, Pal M. Andrographolide upregulates protein quality control mechanisms in cell and mouse through upregulation of mTORC1 function. Biochim Biophys Acta Gen Subj 2021; 1865:129885. [PMID: 33639218 DOI: 10.1016/j.bbagen.2021.129885] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Heat shock response (HSR), a component of cellular protein quality control mechanisms, is defective in different neurodegenerative conditions such as Parkinson's disease (PD). Forced upregulation of heat shock factor 1 (HSF1), an HSR master regulator, showed therapeutic promise in PD models. Many of the reported small-molecule HSF1 activators have limited functions. Therefore, identification and understanding the molecular bases of action of new HSF1 activating molecules is necessary. METHOD We used a cell-based reporter system to screen Andrographis paniculata leaf extract to isolate andrographolide as an inducer of HSF1 activity. The andrographolide activity was characterized by analyzing its role in different protein quality control mechanisms. RESULT We find that besides ameliorating the PD in MPTP-treated mice, andrographolide upregulated different machineries controlled by HSF1 and NRF2 in both cell and mouse brain. Andrographolide achieves these functions through mTORC1 activated via p38 MAPK and ERK pathways. NRF2 activation is reflected in the upregulation of proteasome as well as autophagy pathways. We further show that NRF2 activation is mediated through mTORC1 driven phosphorylation of p62/sequestosome 1. Studies with different cell types suggested that andrographolide-mediated induction of ROS level underlies all these activities in agreement with the upregulation of mTORC1 and NRF2-antioxidant pathway in mice. CONCLUSION Andrographolide through upregulating HSF1 activity ameliorates protein aggregation induced cellular toxicity. GENERAL SIGNIFICANCE Our results provide a reasonable basis for use of andrographolide in the therapy regimen for the treatment of PD.
Collapse
Affiliation(s)
- Naibedya Dutta
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Vinod K Nelson
- Pharmacognosy and Phytotherapy Research Laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Hossainoor R Sareng
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Chirantan Majumder
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Subhash C Mandal
- Pharmacognosy and Phytotherapy Research Laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India.
| |
Collapse
|
31
|
Zhao S, Sheng S, Wang Y, Ding L, Xu X, Xia X, Zheng JC. Astrocyte-derived extracellular vesicles: A double-edged sword in central nervous system disorders. Neurosci Biobehav Rev 2021; 125:148-159. [PMID: 33626395 DOI: 10.1016/j.neubiorev.2021.02.027] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 12/28/2020] [Accepted: 02/16/2021] [Indexed: 12/14/2022]
Abstract
Recent studies suggest that astrocytes released a great quantity of extracellular vesicles (AEVs) to communicate with other brain cells. Under pathological conditions, AEVs are widely associated with the pathogenesis of neurobiological diseases by horizontally transferring pathogenic factors to neighboring cells or peripheral immune cells. Their beneficial role is also evident by the fact that they are involved in neuroprotection and neuroregeneration through alleviating apoptosis, maintaining neuronal function, and repairing neural injuries. The strong association of AEVswith neurological disorders makes AEVs a promising target for disease diagnosis, treatment, and prevention. The identification of disease-specific cargos in AEVs isolated from the patients' biofluids suggests AEVs as an attractive platform for biomarker development. Furthermore, the inhibition of inflammatory/toxic AEV release and the preservation of neuroprotective AEV release have been considered as potential therapeutic strategies in CNS disorder treatment and prevention, respectively. Here, we summarize the biological roles of AEVs as pathological contributors, protective/regenerative factors, and potential diagnostic biomarkers and therapeutic targets for neurological disorders, with a focus on recent progresses and emerging concepts.
Collapse
Affiliation(s)
- Shu Zhao
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Shiyang Sheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Yi Wang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Lu Ding
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaonan Xu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China; Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200434, China.
| | - Jialin C Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China; Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200434, China; Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China; Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930, USA.
| |
Collapse
|
32
|
TRPM2 channel in oxidative stress-induced mitochondrial dysfunction and apoptotic cell death. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 125:51-72. [PMID: 33931144 DOI: 10.1016/bs.apcsb.2020.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mitochondria, conserved intracellular organelles best known as the powerhouse of cells for generating ATP, play an important role in apoptosis. Oxidative stress can induce mitochondrial dysfunction and activate mitochondria-mediated apoptotic cell death. TRPM2 is a Ca2+-permeable cation channel that is activated by pathologically relevant concentrations of reactive oxygen species (ROS) and one of its well-recognized roles is to confer susceptibility to ROS-induced cell death. Increasing evidence from recent studies supports TRPM2 channel-mediated cell death as an important cellular mechanism linking miscellaneous oxidative stress-inducing pathological factors to associated diseased conditions. In this chapter, we will discuss the role of the TRPM2 channel in neurons in the brain and pancreatic β-cells in mediating mitochondrial dysfunction and cell death, focusing mainly on apoptotic cell death, that are induced by pathological stimuli implicated in the pathogenesis of neurodegenerative diseases, ischemic stroke and diabetes.
Collapse
|
33
|
Wang DX, Yang Y, Huang XS, Tang JY, Zhang X, Huang HX, Zhou B, Liu B, Xiao HQ, Li XH, Yang P, Zou SC, Liu K, Wang XY, Li XS. Pramipexole attenuates neuronal injury in Parkinson's disease by targeting miR-96 to activate BNIP3-mediated mitophagy. Neurochem Int 2021; 146:104972. [PMID: 33493581 DOI: 10.1016/j.neuint.2021.104972] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Parkinson's disease is a common neurodegenerative problem. Pramipexole (PPX) plays protective role in Parkinson's disease. Nevertheless, the mechanism of PPX in Parkinson's disease-like neuronal injury is largely uncertain. METHODS 1-methyl-4-phenylpyridinium (MPP+)-stimulated neuronal cells and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mice were used as the model of Parkinson's disease. MPP+-induced neuronal injury was assessed via cell viability, lactic dehydrogenase (LDH) release and apoptosis. microRNA-96 (miR-96) and BCL2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3) abundances were examined by quantitative reverse transcription polymerase chain reaction (qRT-PCR) or Western blotting. Mitophagy was tested by Western blotting and immunofluorescence staining. MPTP-induced neuronal injury in mice was investigated via behavioral tests and TUNEL. RESULTS PPX alleviated MPP+-induced neuronal injury via increasing cell viability and decreasing LDH release and apoptosis. PPX reversed MPP+-induced miR-96 expression and inhibition of mitophagy. miR-96 overexpression or BNIP3 interference weakened the suppressive role of PPX in MPP+-induced neuronal injury. miR-96 targeted BNIP3 to inhibit PTEN-induced putative kinase 1 (PINK1)/Parkin signals-mediated mitophagy. miR-96 overexpression promoted MPP+-induced neuronal injury via decreasing BNIP3. PPX weakened MPTP-induced neuronal injury in mice via regulating miR-96/BNIP3-mediated mitophagy. CONCLUSION PPX mitigated neuronal injury in MPP+-induced cells and MPTP-induced mice by activating BNIP3-mediated mitophagy via directly decreasing miR-96.
Collapse
Affiliation(s)
- Dong-Xin Wang
- The Research Institute of Mentality and Sanitation, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Ying Yang
- Medical Department of Neurology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Xiao-Song Huang
- Medical Department of Neurology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Jia-Yu Tang
- Medical Department of Neurology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Xi Zhang
- Clinical Medical School, Hunan Traditional Chinese Medicine University, Changsha, 410006, Hunan Province, PR China
| | - Hong-Xing Huang
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Bin Zhou
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Bo Liu
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Hui-Qiong Xiao
- Department of Scientific Research, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Xiao-Hui Li
- Medical Department of Neurology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Ping Yang
- Department of Psychology, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Shu-Cheng Zou
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Kun Liu
- Department of Neurosurgery, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China
| | - Xiao-Ye Wang
- The Institution of Clinical Trials on Drugs, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China.
| | - Xiao-Song Li
- The Research Institute of Mentality and Sanitation, Hunan Provincial Brain Hospital, Changsha, 410007, Hunan Province, PR China.
| |
Collapse
|
34
|
Yuan LJ, Zhang M, Chen S, Chen WF. Anti-inflammatory effect of IGF-1 is mediated by IGF-1R cross talk with GPER in MPTP/MPP +-induced astrocyte activation. Mol Cell Endocrinol 2021; 519:111053. [PMID: 33035625 DOI: 10.1016/j.mce.2020.111053] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 12/17/2022]
Abstract
Insulin-like growth factor-1 (IGF-1) is a potent neuroprotective polypeptide that exerts neuroprotective effects via the IGF-1 receptor (IGF-1R). Our previous study reported that G protein-coupled estrogen receptor (GPER) was involved in the anti-apoptotic effect of IGF-1. The present study was designed to investigate the anti-inflammatory effect of IGF-1 in association with astrocyte activation and the molecular details of the interaction between IGF-1R and GPER. We showed that IGF-1 could improve 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced motor deficits and attenuate the upregulation of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) both in vivo and in vitro. The IGF-1R antagonist JB-1 and the GPER antagonist G15 could antagonize the anti-inflammatory effect of IGF-1. Silencing GPER abrogated the inhibitory effect of IGF-1 on 1-methyl-4-phenylpyridinium (MPP+)-induced upregulation of COX-2 and iNOS in primary astrocytes. Moreover, the MPP + -induced inflammatory response was related to the activation of mitogen-activated protein kinases (MAPKs) and NF-κB signaling pathways. The inhibitory effects of IGF-1 on the phosphorylation of p38, JNK and IκB could be blocked by JB-1. G15 antagonized the inhibitory effects of IGF-1 on p-JNK and p-IκB, but not p-p38. Furthermore, IGF-1 treatment alone increased the expression of GPER, which was blocked by JB-1, the phosphatidylinositol 3-kinase (PI3-K) antagonist LY294002 and the MEK antagonist PD98059 in primary astrocytes. Overall, we show for the first time that GPER may contribute to the anti-inflammatory effect of IGF-1 against MPTP/MPP + -induced astrocyte activation. IGF-1 could regulate the expression of GPER via the IGF-1R/PI3-K/MAPK signaling pathway in primary astrocytes.
Collapse
MESH Headings
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- 1-Methyl-4-phenylpyridinium/toxicity
- Animals
- Anti-Inflammatory Agents/pharmacology
- Astrocytes/drug effects
- Astrocytes/metabolism
- Behavior, Animal
- Benzodioxoles/pharmacology
- Cells, Cultured
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Humans
- Insulin-Like Growth Factor I/pharmacology
- MAP Kinase Signaling System/drug effects
- Male
- Mice, Inbred C57BL
- NF-kappa B/metabolism
- Nitric Oxide Synthase Type II/genetics
- Nitric Oxide Synthase Type II/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation/drug effects
- Quinolines/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, IGF Type 1/metabolism
- Receptors, Estrogen/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Substantia Nigra/metabolism
- Up-Regulation/drug effects
- Mice
Collapse
Affiliation(s)
- Liang-Jie Yuan
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China; School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Mei Zhang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Su Chen
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Wen-Fang Chen
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
35
|
Mao Z, Wen-Ting Z, Hai-Tao W, Hui Y, Shi-Yi L, Jiang-Ping X, Wen-Ya W. AMI, an Indazole Derivative, Improves Parkinson's Disease by Inhibiting Tau Phosphorylation. Front Mol Neurosci 2020; 13:165. [PMID: 33328879 PMCID: PMC7710523 DOI: 10.3389/fnmol.2020.00165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/11/2020] [Indexed: 11/17/2022] Open
Abstract
Dopaminergic neuronal loss is the main pathological character of Parkinson’s disease (PD). Abnormal tau hyperphosphorylation will lead to dopaminergic neuronal loss. An indazole derivative 6-amino-1-methyl-indazole (AMI) successfully synthesized to inhibit tau hyperphosphorylation may exert a neuroprotective effect. The in vitro study showed that AMI effectively increased cell viability and alleviated the apoptosis induced by MPP+ in SH-SY5Y cells. In addition, AMI treatment significantly decreased the expression of p-tau and upstream kinases GSK-3β. In the MPTP-induced PD mice models, we found AMI apparently preserved dopaminergic neurons in the substantia nigra and improved the PD behavioral symptoms. Our results demonstrate that AMI exerts a neuroprotective effect by inhibiting tau hyperphosphorylation, representing a promising new candidate for PD treatment.
Collapse
Affiliation(s)
- Zhang Mao
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Zhu Wen-Ting
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wang Hai-Tao
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yu Hui
- School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Lan Shi-Yi
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xu Jiang-Ping
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wang Wen-Ya
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
36
|
Meng C, Gao J, Ma Q, Sun Q, Qiao T. LINC00943 knockdown attenuates MPP +-induced neuronal damage via miR-15b-5p/RAB3IP axis in SK-N-SH cells. Neurol Res 2020; 43:181-190. [PMID: 33208053 DOI: 10.1080/01616412.2020.1834290] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Parkinson's disease (PD) is a neurodegenerative problem correlated with neuronal damage. Long noncoding RNAs (lncRNAs) are implicated in neuronal damage in PD development. This research aims to analyze the function and mechanism of LINC00943 in 1-methyl-4-phenylpyridinium (MPP+)-caused neuronal injury. METHODS MPP+-challenged SK-N-SH cells served as a PD-like model of neuronal damage. LINC00943, microRNA-15b-5p (miR-15b-5p) and RAB3A interacting protein (RAB3IP) abundances were examined via quantitative reverse transcription polymerase chain reaction or western blot. MPP+-caused neuronal damage was assessed via cell viability, apoptosis, inflammatory injury and oxidative injury. The association between miR-15b-5p and LINC00943 or RAB3IP was determined via dual-luciferase reporter analysis and RNA immunoprecipitation. RESULTS LINC00943 abundance was up-regulated in MPP+-challenged SK-N-SH cells. LINC00943 silence alleviated MPP+-caused decrease of cell viability and elevation of apoptosis, inflammatory injury and oxidative injury. miR-15b-5p was inhibited via LINC00943, and miR-15b-5p inhibition reversed knockdown of LINC00943-mediated suppression of MPP+-induced neuronal damage. RAB3IP was targeted via miR-15b-5p, and LINC00943 could regulate RAB3IP via miR-15b-5p. miR-15b-5p addition mitigated MPP+-induced neuronal damage through decreasing RAB3IP. CONCLUSION LINC00943 inhibition alleviated MPP+-induced neuronal injury via miR-15b-5p/RAB3IP axis, indicating a potential target for treatment of PD.
Collapse
Affiliation(s)
- Chunming Meng
- Department of Geriatrics, The First People's Hospital of Lianyungang , Lianyungan City, Jiangsu Province, China
| | - Jing Gao
- Department of Geriatrics, The First People's Hospital of Lianyungang , Lianyungan City, Jiangsu Province, China
| | - Qingyuan Ma
- Department of Geriatrics, The First People's Hospital of Lianyungang , Lianyungan City, Jiangsu Province, China
| | - Qian Sun
- Department of Geriatrics, The First People's Hospital of Lianyungang , Lianyungan City, Jiangsu Province, China
| | - Tiantian Qiao
- Department of Geriatrics, The First People's Hospital of Lianyungang , Lianyungan City, Jiangsu Province, China
| |
Collapse
|
37
|
Molecular Mechanism of Platelet-Derived Growth Factor (PDGF)-BB-Mediated Protection Against MPP+ Toxicity in SH-SY5Y Cells. J Mol Neurosci 2020; 71:1131-1143. [DOI: 10.1007/s12031-020-01735-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 10/02/2020] [Indexed: 12/21/2022]
|
38
|
Mir-141-3p Regulates Apoptosis and Mitochondrial Membrane Potential via Targeting Sirtuin1 in a 1-Methyl-4-Phenylpyridinium in vitro Model of Parkinson's Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7239895. [PMID: 33204711 PMCID: PMC7666638 DOI: 10.1155/2020/7239895] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/24/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
Objectives Parkinson's disease (PD) is a common neurodegenerative disease characterized by the loss of midbrain dopaminergic neurons in the substantia nigra. The present study investigated miR-141-3p/sirtuin1 (SIRT1) activity in a 1-methyl-4-phenylpyridinium- (MPP+-) induced PC12-cell model of PD. Methods PC12 cells were exposed to MMP+ following induction of differentiation by nerve growth factor (NGF). miR-141-3p and SIRT1 expressions were examined using RT-qPCR and western blot. Cell viability was evaluated using the MTT assay. Apoptosis percentage, reactive oxygen species (ROS) production, and mitochondrial membrane potential (Δψm) were evaluated using flow cytometry. Expression of Nuclear factor-kappa B- (NF-κB-) related proteins was determined by western blot. Bioinformatic analysis, RT-qPCR, and luciferase reporter assay were used to confirm the interaction between miR-141-3p and SIRT1. Results miR-141-3p was upregulated, and SIRT1 was downregulated in MPP+-treated PC12 cells. MPP+ treatment also upregulated nitric oxide synthase 1 (Nos1) and α-synuclein. miR-141-3p induced apoptosis, oxidative stress, mitochondrial dysfunction, and downregulated the SIRT1 mRNA expression. The luciferase reporter assay showed that SIRT1 was the target of miR-141-3p. SIRT1 transfection attenuated apoptosis, ROS production and maintained Δψm. SIRT1 also downregulated Nos1, tumor necrosis factor-α (TNF-α), interleukin 1 beta (IL-1β), interleukin 6(IL-6) and upregulated B cell lymphoma 2 (Bcl-2) protein. In addition, SIRT1 activator resveratrol blocked the effects of miR-141-3p mimic on Nos1, α-synuclein, and mitochondrial membrane potential. SIRT1 inhibitor sirtinol reversed the biological effects of miR-141-3p. Conclusion Increased miR-141-3p induced apoptosis, oxidative stress, and mitochondrial dysfunction in MPP+-treated PC12 cells by directly targeting the SIRT1 expression. Our study provided a potential therapeutic strategy for PD.
Collapse
|
39
|
Malko P, Jiang LH. TRPM2 channel-mediated cell death: An important mechanism linking oxidative stress-inducing pathological factors to associated pathological conditions. Redox Biol 2020; 37:101755. [PMID: 33130440 PMCID: PMC7600390 DOI: 10.1016/j.redox.2020.101755] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/17/2020] [Accepted: 10/08/2020] [Indexed: 12/26/2022] Open
Abstract
Oxidative stress resulting from the accumulation of high levels of reactive oxygen species is a salient feature of, and a well-recognised pathological factor for, diverse pathologies. One common mechanism for oxidative stress damage is via the disruption of intracellular ion homeostasis to induce cell death. TRPM2 is a non-selective Ca2+-permeable cation channel with a wide distribution throughout the body and is highly sensitive to activation by oxidative stress. Recent studies have collected abundant evidence to show its important role in mediating cell death induced by miscellaneous oxidative stress-inducing pathological factors, both endogenous and exogenous, including ischemia/reperfusion and the neurotoxicants amyloid-β peptides and MPTP/MPP+ that cause neuronal demise in the brain, myocardial ischemia/reperfusion, proinflammatory mediators that disrupt endothelial function, diabetogenic agent streptozotocin and diabetes risk factor free fatty acids that induce loss of pancreatic β-cells, bile acids that damage pancreatic acinar cells, renal ischemia/reperfusion and albuminuria that are detrimental to kidney cells, acetaminophen that triggers hepatocyte death, and nanoparticles that injure pericytes. Studies have also shed light on the signalling mechanisms by which these pathological factors activate the TRPM2 channel to alter intracellular ion homeostasis leading to aberrant initiation of various cell death pathways. TRPM2-mediated cell death thus emerges as an important mechanism in the pathogenesis of conditions including ischemic stroke, neurodegenerative diseases, cardiovascular diseases, diabetes, pancreatitis, chronic kidney disease, liver damage and neurovascular injury. These findings raise the exciting perspective of targeting the TRPM2 channel as a novel therapeutic strategy to treat such oxidative stress-associated diseases.
Collapse
Affiliation(s)
- Philippa Malko
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province and Department of Physiology and Pathophysiology, Xinxiang Medical University, PR China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK.
| |
Collapse
|
40
|
Design and evaluation of bi-functional iron chelators for protection of dopaminergic neurons from toxicants. Arch Toxicol 2020; 94:3105-3123. [PMID: 32607613 PMCID: PMC7415766 DOI: 10.1007/s00204-020-02826-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/24/2020] [Indexed: 02/07/2023]
Abstract
While the etiology of non-familial Parkinson’s disease (PD) remains unclear, there is evidence that increased levels of tissue iron may be a contributing factor. Moreover, exposure to some environmental toxicants is considered an additional risk factor. Therefore, brain-targeted iron chelators are of interest as antidotes for poisoning with dopaminergic toxicants, and as potential treatment of PD. We, therefore, designed a series of small molecules with high affinity for ferric iron and containing structural elements to allow their transport to the brain via the neutral amino acid transporter, LAT1 (SLC7A5). Five candidate molecules were synthesized and initially characterized for protection from ferroptosis in human neurons. The promising hydroxypyridinone SK4 was characterized further. Selective iron chelation within the physiological range of pH values and uptake by LAT1 were confirmed. Concentrations of 10–20 µM blocked neurite loss and cell demise triggered by the parkinsonian neurotoxicants, methyl-phenyl-pyridinium (MPP+) and 6-hydroxydopamine (6-OHDA) in human dopaminergic neuronal cultures (LUHMES cells). Rescue was also observed when chelators were given after the toxicant. SK4 derivatives that either lacked LAT1 affinity or had reduced iron chelation potency showed altered activity in our assay panel, as expected. Thus, an iron chelator was developed that revealed neuroprotective properties, as assessed in several models. The data strongly support the role of iron in dopaminergic neurotoxicity and suggests further exploration of the proposed design strategy for improving brain iron chelation.
Collapse
|
41
|
Shakespear N, Ogura M, Yamaki J, Homma Y. Astrocyte-Derived Exosomal microRNA miR-200a-3p Prevents MPP +-Induced Apoptotic Cell Death Through Down-Regulation of MKK4. Neurochem Res 2020; 45:1020-1033. [PMID: 32016794 DOI: 10.1007/s11064-020-02977-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/06/2019] [Accepted: 01/26/2020] [Indexed: 02/07/2023]
Abstract
Astrocytes release exosomes that regulate neuronal cell function. 1-methyl-4-phenylpyridinium (MPP+) is a well-known neurotoxin used to induce cell death in in vitro Parkinson's disease models, and microRNA (miRNA) transferred by released exosomes can regulate its mechanisms. Here, we demonstrated that exosomes released from normal astrocytes (ADEXs), but not exosomes derived from MPP+-stimulated astrocytes (MPP+-ADEXs), significantly attenuate MPP+-induced cell death in SH-SY5Y cells and primary mesencephalic dopaminergic neuron cultures, and reduce expression of mitogen-activated protein kinase kinase 4 (MKK4), an important upstream kinase in the c-Jun N-terminal kinase cell death pathway. Similar neuroprotective results were obtained from primary hippocampal neuron cultures, an in vitro glutamate excitotoxicity model. Through small-RNA sequencing of exosomal miRNA, we identified miR-200a-3p as the most down-regulated miRNA expressed in MPP+-ADEXs. miRNA target analysis and reporter assay confirmed that miR-200a-3p targets MKK4 through binding to two independent sites on the 3'-UTR of Map2k4/MKK4 mRNA. Treatment with miR-200a-3p mimic suppressed both MKK4 mRNA and protein expressions, and attenuated cell death in MPP+-treated SH-SY5Y cells and glutamate-treated hippocampal neuron cultures. Our results suggest that normal astrocytes release miR-200a-3p which exhibits a neuroprotective effect through down-regulation of MKK4.
Collapse
Affiliation(s)
| | - Masato Ogura
- Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Junko Yamaki
- Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Yoshimi Homma
- Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan.
| |
Collapse
|
42
|
Lu J, Dou F, Yu Z. The potassium channel KCa3.1 represents a valid pharmacological target for microgliosis-induced neuronal impairment in a mouse model of Parkinson's disease. J Neuroinflammation 2019; 16:273. [PMID: 31878950 PMCID: PMC6931251 DOI: 10.1186/s12974-019-1682-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Background Recent studies described a critical role for microglia in Parkinson’s disease (PD), where these central nerve system resident immune cells participate in the neuroinflammatory microenvironment that contributes to dopaminergic neurons loss in the substantia nigra. Understanding the phenotype switch of microgliosis in PD could help to identify the molecular mechanism which could attenuate or delay the progressive decline in motor function. KCa3.1 has been reported to regulate the “pro-inflammatory” phenotype switch of microglia in neurodegenerative pathological conditions. Methods We here investigated the effects of gene deletion or pharmacological blockade of KCa3.1 activity in wild-type or KCa3.1−/− mice after treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a mouse model of PD. MPTP-induced PD mouse model was subjected to the rotarod test to evaluate the locomotor ability. Glia activation and neuron loss were measured by immunostaining. Fluo-4 AM was used to measure cytosolic Ca2+ level in 1-methyl-4-phenylpyridinium (MPP+)-induced microgliosis in vitro. Results We report that treatment of MPTP-induced PD mouse model with gene deletion or pharmacological blockade of KCa3.1 with senicapoc improves the locomotor ability and the tyrosine hydroxylase (TH)-positive neuron number and attenuates the microgliosis and neuroinflammation in the substantia nigra pars compacta (SNpc). KCa3.1 involves in store-operated Ca2+ entry-induced Ca2+ overload and endoplasmic reticulum stress via the protein kinase B (AKT) signaling pathway during microgliosis. Gene deletion or blockade of KCa3.1 restored AKT/mammalian target of rapamycin (mTOR) signaling both in vivo and in vitro. Conclusions Taken together, these results demonstrate a key role for KCa3.1 in driving a pro-inflammatory microglia phenotype in PD.
Collapse
Affiliation(s)
- Jia Lu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Fangfang Dou
- Basic Research Department, Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China
| | - Zhihua Yu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
43
|
Selective vulnerability in α-synucleinopathies. Acta Neuropathol 2019; 138:681-704. [PMID: 31006067 PMCID: PMC6800835 DOI: 10.1007/s00401-019-02010-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/13/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022]
Abstract
Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy are neurodegenerative disorders resulting in progressive motor/cognitive deficits among other symptoms. They are characterised by stereotypical brain cell loss accompanied by the formation of proteinaceous aggregations of the protein α-synuclein (α-syn), being, therefore, termed α-synucleinopathies. Although the presence of α-syn inclusions is a common hallmark of these disorders, the exact nature of the deposited protein is specific to each disease. Different neuroanatomical regions and cellular populations manifest a differential vulnerability to the appearance of protein deposits, cell dysfunction, and cell death, leading to phenotypic diversity. The present review describes the multiple factors that contribute to the selective vulnerability in α-synucleinopathies. We explore the intrinsic cellular properties in the affected regions, including the physiological and pathophysiological roles of endogenous α-syn, the metabolic and genetic build-up of the cells and their connectivity. These factors converge with the variability of the α-syn conformational strains and their spreading capacity to dictate the phenotypic diversity and regional vulnerability of each disease. Finally, we describe the exogenous and environmental factors that potentially contribute by igniting and modulating the differential pathology in α-synucleinopathies. In conclusion, we think that it is the confluence of this disruption of the cellular metabolic state and α-syn structural equilibrium through the anatomical connectivity which appears to initiate cascades of pathological processes triggered by genetic, environmental, or stochastic events that result in the "death by a thousand cuts" profile of α-synucleinopathies.
Collapse
|
44
|
Delp J, Funke M, Rudolf F, Cediel A, Bennekou SH, van der Stel W, Carta G, Jennings P, Toma C, Gardner I, van de Water B, Forsby A, Leist M. Development of a neurotoxicity assay that is tuned to detect mitochondrial toxicants. Arch Toxicol 2019; 93:1585-1608. [PMID: 31190196 DOI: 10.1007/s00204-019-02473-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/07/2019] [Indexed: 12/18/2022]
Abstract
Many neurotoxicants affect energy metabolism in man, but currently available test methods may still fail to predict mito- and neurotoxicity. We addressed this issue using LUHMES cells, i.e., human neuronal precursors that easily differentiate into mature neurons. Within the NeuriTox assay, they have been used to screen for neurotoxicants. Our new approach is based on culturing the cells in either glucose or galactose (Glc-Gal-NeuriTox) as the main carbohydrate source during toxicity testing. Using this Glc-Gal-NeuriTox assay, 52 mitochondrial and non-mitochondrial toxicants were tested. The panel of chemicals comprised 11 inhibitors of mitochondrial respiratory chain complex I (cI), 4 inhibitors of cII, 8 of cIII, and 2 of cIV; 8 toxicants were included as they are assumed to be mitochondrial uncouplers. In galactose, cells became more dependent on mitochondrial function, which made them 2-3 orders of magnitude more sensitive to various mitotoxicants. Moreover, galactose enhanced the specific neurotoxicity (destruction of neurites) compared to a general cytotoxicity (plasma membrane lysis) of the toxicants. The Glc-Gal-NeuriTox assay worked particularly well for inhibitors of cI and cIII, while the toxicity of uncouplers and non-mitochondrial toxicants did not differ significantly upon glucose ↔ galactose exchange. As a secondary assay, we developed a method to quantify the inhibition of all mitochondrial respiratory chain functions/complexes in LUHMES cells. The combination of the Glc-Gal-NeuriTox neurotoxicity screening assay with the mechanistic follow up of target site identification allowed both, a more sensitive detection of neurotoxicants and a sharper definition of the mode of action of mitochondrial toxicants.
Collapse
Affiliation(s)
- Johannes Delp
- Chair for In Vitro Toxicology and Biomedicine, Department of Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
- Cooperative Doctorate College InViTe, University of Konstanz, Constance, Germany
| | - Melina Funke
- Chair for In Vitro Toxicology and Biomedicine, Department of Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Franziska Rudolf
- Chair for In Vitro Toxicology and Biomedicine, Department of Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - Andrea Cediel
- Swetox Unit for Toxicological Sciences, Karolinska Institutet, Stockholm, Sweden
| | | | - Wanda van der Stel
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Giada Carta
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Cosimo Toma
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156, Milan, Italy
| | | | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Anna Forsby
- Swetox Unit for Toxicological Sciences, Karolinska Institutet, Stockholm, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Marcel Leist
- Chair for In Vitro Toxicology and Biomedicine, Department of Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany.
| |
Collapse
|
45
|
He X, Yang S, Zhang R, Hou L, Xu J, Hu Y, Xu R, Wang H, Zhang Y. Smilagenin Protects Dopaminergic Neurons in Chronic MPTP/Probenecid-Lesioned Parkinson's Disease Models. Front Cell Neurosci 2019; 13:18. [PMID: 30804756 PMCID: PMC6371654 DOI: 10.3389/fncel.2019.00018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/16/2019] [Indexed: 01/22/2023] Open
Abstract
Current therapies for Parkinson’s disease (PD) only offer limited symptomatic alleviation but fail to hamper the progress of the disease. Thus, it is imperative to establish new approaches aiming at protecting or reversing neurodegeneration in PD. Recent work elucidates whether smilagenin (abbreviated SMI), a steroidal sapogenin from traditional Chinese medicinal herbs, can take neuroprotective effect on dopaminergic neurons in a chronic model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) conjuncted with probenecid mice. We reported for the first time that SMI significantly improved the locomotor ability of chronic MPTP/probenecid–lesioned mice. SMI increased the tyrosine hydroxylase (TH) positive and Nissl positive neuron number in the substantia nigra pars compacta (SNpc), augmented striatal DA and its metabolites concentration and elevated striatal dopamine transporter density (DAT). In addition, dopamine receptor D2R not D1R was down-regulated by MPTP/probenecid and slightly raised by SMI prevention. What’s more, we discovered that SMI markedly elevated striatal glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) protein levels in SMI prevented mice. And we found that SMI increased GDNF and BDNF mRNA level by promoting CREB phosphorylation in 1-methyl-4-phenylpyridimium (MPP+) treated SH-SY5Y cells. The results illustrated that SMI could prevent the impairment of dopaminergic neurons in chronic MPTP/probenecid-induced mouse model.
Collapse
Affiliation(s)
- Xuan He
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Shuangshuang Yang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Rui Zhang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Lina Hou
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Jianrong Xu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Yaer Hu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Rang Xu
- Scientific Research Center, Xinhua Hospital, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Hao Wang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| | - Yongfang Zhang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai JiaoTong University School of Medicine (SJTUSM), Shanghai, China
| |
Collapse
|
46
|
Liss B, Striessnig J. The Potential of L-Type Calcium Channels as a Drug Target for Neuroprotective Therapy in Parkinson's Disease. Annu Rev Pharmacol Toxicol 2019; 59:263-289. [PMID: 30625283 DOI: 10.1146/annurev-pharmtox-010818-021214] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The motor symptoms of Parkinson's disease (PD) mainly arise from degeneration of dopamine neurons within the substantia nigra. As no disease-modifying PD therapies are available, and side effects limit long-term benefits of current symptomatic therapies, novel treatment approaches are needed. The ongoing phase III clinical study STEADY-PD is investigating the potential of the dihydropyridine isradipine, an L-type Ca2+ channel (LTCC) blocker, for neuroprotective PD therapy. Here we review the clinical and preclinical rationale for this trial and discuss potential reasons for the ambiguous outcomes of in vivo animal model studies that address PD-protective dihydropyridine effects. We summarize current views about the roles of Cav1.2 and Cav1.3 LTCC isoforms for substantia nigra neuron function, and their high vulnerability to degenerative stressors, and for PD pathophysiology. We discuss different dihydropyridine sensitivities of LTCC isoforms in view of their potential as drug targets for PD neuroprotection, and we conclude by considering how these aspects could guide further drug development.
Collapse
Affiliation(s)
- Birgit Liss
- Institut für Angewandte Physiologie, Universität Ulm, 89081 Ulm, Germany;
| | - Jörg Striessnig
- Abteilung Pharmakologie und Toxikologie, Institut für Pharmazie, and Center for Molecular Biosciences Innsbruck, Universität Innsbruck, A-6020 Innsbruck, Austria;
| |
Collapse
|
47
|
Gutbier S, Spreng AS, Delp J, Schildknecht S, Karreman C, Suciu I, Brunner T, Groettrup M, Leist M. Prevention of neuronal apoptosis by astrocytes through thiol-mediated stress response modulation and accelerated recovery from proteotoxic stress. Cell Death Differ 2018; 25:2101-2117. [PMID: 30390092 PMCID: PMC6261954 DOI: 10.1038/s41418-018-0229-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 09/12/2018] [Accepted: 10/11/2018] [Indexed: 01/04/2023] Open
Abstract
The development of drugs directly interfering with neurodegeneration has proven to be astonishingly difficult. Alternative therapeutic approaches could result from a better understanding of the supportive function of glial cells for stressed neurons. Therefore, here, we investigated the mechanisms involved in the endogenous neuro-defensive activity of astrocytes. A well-established model of postmitotic human dopaminergic neurons (LUHMES cells) was used in the absence ('LUHMES' mono-culture) or presence ('co-culture') of astrocytes. Inhibition of the LUHMES proteasome led to proteotoxic (protein aggregates; ATF-4 induction) and oxidative (GSH-depletion; NRF-2 induction) stress, followed by neuronal apoptosis. The presence of astrocytes attenuated the neuronal stress response, and drastically reduced neurodegeneration. A similar difference between LUHMES mono- and co-cultures was observed, when proteotoxic and oxidative stress was triggered indirectly by inhibitors of mitochondrial function (rotenone, MPP+). Human and murine astrocytes continuously released glutathione (GSH) into the medium, and transfer of glia-conditioned medium was sufficient to rescue LUHMES, unless it was depleted for GSH. Also, direct addition of GSH to LUHMES rescued the neurons from inhibition of the proteasome. Both astrocytes and GSH blunted the neuronal ATF-4 response and similarly upregulated NRF-1/NFE2L1, a transcription factor counter-regulating neuronal proteotoxic stress. Astrocyte co-culture also helped to recover the neurons' ability to degrade aggregated poly-ubiquitinated proteins. Overexpression of NRF-1 attenuated the toxicity of proteasome inhibition, while knockdown increased toxicity. Thus, astrocytic thiol supply increased neuronal resilience to various proteotoxic stressors by simultaneously attenuating cell death-related stress responses, and enhancing the recovery from proteotoxic stress through upregulation of NRF-1.
Collapse
Affiliation(s)
- Simon Gutbier
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
- Research Training Group RTG1331, University of Konstanz, Konstanz, Germany
| | - Anna-Sophie Spreng
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Constance, Germany
| | - Johannes Delp
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
- Research Training Group RTG1331, University of Konstanz, Konstanz, Germany
- Cooperative Doctorate College InViTe, University of Konstanz, Konstanz, Germany
| | - Stefan Schildknecht
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Christiaan Karreman
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Ilinca Suciu
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Constance, Germany
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Marcus Groettrup
- Division of Immunology, Department of Biology, University of Konstanz, D-78457, Konstanz, Germany
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany.
- CAAT-Europe, University of Konstanz, 78457, Konstanz, Germany.
| |
Collapse
|
48
|
Major changes of cell function and toxicant sensitivity in cultured cells undergoing mild, quasi-natural genetic drift. Arch Toxicol 2018; 92:3487-3503. [PMID: 30298209 PMCID: PMC6290691 DOI: 10.1007/s00204-018-2326-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022]
Abstract
Genomic drift affects the functional properties of cell lines, and the reproducibility of data from in vitro studies. While chromosomal aberrations and mutations in single pivotal genes are well explored, little is known about effects of minor, possibly pleiotropic, genome changes. We addressed this question for the human dopaminergic neuronal precursor cell line LUHMES by comparing two subpopulations (SP) maintained either at the American-Type-Culture-Collection (ATCC) or by the original provider (UKN). Drastic differences in susceptibility towards the specific dopaminergic toxicant 1-methyl-4-phenylpyridinium (MPP+) were observed. Whole-genome sequencing was performed to identify underlying genetic differences. While both SP had normal chromosome structures, they displayed about 70 differences on the level of amino acid changing events. Some of these differences were confirmed biochemically, but none offered a direct explanation for the altered toxicant sensitivity pattern. As second approach, markers known to be relevant for the intended use of the cells were specifically tested. The “ATCC” cells rapidly down-regulated the dopamine-transporter and tyrosine-hydroxylase after differentiation, while “UKN” cells maintained functional levels. As the respective genes were not altered themselves, we conclude that polygenic complex upstream changes can have drastic effects on biochemical features and toxicological responses of relatively similar SP of cells.
Collapse
|
49
|
Kilbourn MR, Scott PJH. Issues in preclinical radiopharmaceutical research: Significance, relevance and reproducibility. Nucl Med Biol 2018; 67:52-55. [PMID: 30209016 DOI: 10.1016/j.nucmedbio.2018.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/02/2018] [Accepted: 07/16/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Michael R Kilbourn
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI 48105, United States of America.
| | - Peter J H Scott
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI 48105, United States of America
| |
Collapse
|
50
|
Harris G, Eschment M, Orozco SP, McCaffery JM, Maclennan R, Severin D, Leist M, Kleensang A, Pamies D, Maertens A, Hogberg HT, Freeman D, Kirkwood A, Hartung T, Smirnova L. Toxicity, recovery, and resilience in a 3D dopaminergic neuronal in vitro model exposed to rotenone. Arch Toxicol 2018; 92:2587-2606. [PMID: 29955902 PMCID: PMC6063347 DOI: 10.1007/s00204-018-2250-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023]
Abstract
To date, most in vitro toxicity testing has focused on acute effects of compounds at high concentrations. This testing strategy does not reflect real-life exposures, which might contribute to long-term disease outcome. We used a 3D-human dopaminergic in vitro LUHMES cell line model to determine whether effects of short-term rotenone exposure (100 nM, 24 h) are permanent or reversible. A decrease in complex I activity, ATP, mitochondrial diameter, and neurite outgrowth were observed acutely. After compound removal, complex I activity was still inhibited; however, ATP levels were increased, cells were electrically active and aggregates restored neurite outgrowth integrity and mitochondrial morphology. We identified significant transcriptomic changes after 24 h which were not present 7 days after wash-out. Our results suggest that testing short-term exposures in vitro may capture many acute effects which cells can overcome, missing adaptive processes, and long-term mechanisms. In addition, to study cellular resilience, cells were re-exposed to rotenone after wash-out and recovery period. Pre-exposed cells maintained higher metabolic activity than controls and presented a different expression pattern in genes previously shown to be altered by rotenone. NEF2L2, ATF4, and EAAC1 were downregulated upon single hit on day 14, but unchanged in pre-exposed aggregates. DAT and CASP3 were only altered after re-exposure to rotenone, while TYMS and MLF1IP were downregulated in both single-exposed and pre-exposed aggregates. In summary, our study shows that a human cell-based 3D model can be used to assess cellular adaptation, resilience, and long-term mechanisms relevant to neurodegenerative research.
Collapse
Affiliation(s)
- Georgina Harris
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Melanie Eschment
- Center for Alternatives to Animal Testing (CAAT) Europe, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Sebastian Perez Orozco
- The Integrated Imaging Center, Department of Biology, Engineering in Oncology Center and The Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - J Michael McCaffery
- The Integrated Imaging Center, Department of Biology, Engineering in Oncology Center and The Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Daniel Severin
- The Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Marcel Leist
- Center for Alternatives to Animal Testing (CAAT) Europe, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Andre Kleensang
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - David Pamies
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alexandra Maertens
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Dana Freeman
- Department of Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alfredo Kirkwood
- The Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA.,Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Center for Alternatives to Animal Testing (CAAT) Europe, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|