1
|
Gamit N, Patil M, Soumya BS, Dharmarajan A, Warrier S. Development of In Vitro Parkinson's Disease Model Mediated by MPP+ and α-Synuclein Using Wharton's Jelly Mesenchymal Stem Cells. CNS Neurosci Ther 2025; 31:e70299. [PMID: 40260646 PMCID: PMC12012574 DOI: 10.1111/cns.70299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 04/23/2025] Open
Abstract
MAIN PROBLEM The mechanism behind Parkinson's disease (PD) is still unclear, and a cure to stop its progression is yet to be found. This is mainly due to the lack of effective human PD models. To address this, we generated an in vitro PD model using Wharton's jelly-derived mesenchymal stem cells (WJMSCs). METHODS WJMSCs were isolated from the umbilical cord using an enzymatic method. MSCs were characterized by RT-PCR, immunofluorescence, and trilineage differentiation. MSCs were differentiated into dopaminergic neuron-like cells (DAN) and further degenerated by treating them with either MPP+ iodide or the A53T mutated α-synuclein variant. Gene expression analysis by qRT-PCR and protein analysis by immunofluorescence, flow cytometry, and ELISA were performed. Assays to measure LDH, ROS, NO, GSH, and mitochondrial membrane potential were also performed after degeneration. RESULTS WJMSCs were positive for MSC markers and were able to differentiate into adipocytes, chondrocytes, and osteocytes. DAN obtained after the differentiation of WJMSCs for 48 h expressed neuronal markers such as synapsin 1, neuropilin, neurofilament, and MAPT along with dopaminergic markers such as Nurr1, DAT, TH, DDC, and KCNJ6 and were functionally active. Upon degeneration of DAN by MPP+ or A53T, elevated levels of SNCA and downregulation of TH, Nurr1, DAT, and KCNJ6 were observed. Furthermore, increased expression of α-SYN was detected at the protein level as well. Finally, reduction in mitochondrial membrane potential and GSH levels along with an increase in intracellular ROS, nitrite production, and LDH levels confirmed that the in vitro PD-like model exhibited the molecular characteristics of PD. CONCLUSION This model is rapid, cost-efficient, and effective for understanding the molecular mechanisms of the disease and can also be used for screening of emerging therapeutics for PD.
Collapse
Affiliation(s)
- Naisarg Gamit
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal Academy of Higher Education (MAHE)BangaloreIndia
| | - Manasi Patil
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal Academy of Higher Education (MAHE)BangaloreIndia
| | - B. S. Soumya
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal Academy of Higher Education (MAHE)BangaloreIndia
| | - Arun Dharmarajan
- School of Human SciencesThe University of Western AustraliaNedlandsWestern AustraliaAustralia
- Curtin Medical SchoolCurtin UniversityPerthWestern AustraliaAustralia
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative MedicineManipal Academy of Higher Education (MAHE)BangaloreIndia
- Department of Biotechnology, Faculty of Biomedical Sciences and TechnologySri Ramachandra Institute of Higher Education and ResearchChennaiIndia
| |
Collapse
|
2
|
Liu Y, Cao L, Zhou J, Li C, Du J, Xue Y, Li X, Mao L. Single-Vesicle Electrochemistry in Fresh Brain Slices Enables In Situ Quantification of Vesicular Monoamine. J Am Chem Soc 2025; 147:149-160. [PMID: 39705330 DOI: 10.1021/jacs.4c06572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
The quantitative analysis of vesicular neurotransmitters in neurons in situ is paramount for investigating neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease (PD). Unfortunately, a direct approach for monitoring neurotransmitter chemistry in single vesicles in fresh brain tissue has remained inaccessible so far. Here, we introduce an innovative platform of single-vesicle electrochemistry (SVE) in fresh brain tissue, enabling the quantification of neurotransmitters at the single-vesicle level for both soma and varicosity. Utilizing this methodology, we investigated a PD animal model, which demonstrated a significant reduction in both vesicular dopamine (DA) storage and exocytotic release in DA neurons in the substantia nigra. Furthermore, SVE unveiled the heterogeneous nature of chemical neurotransmission among DA neurons across different brain regions. Importantly, this tissue-based SVE approach can be extended to quantify other monoamine neurotransmitters, such as norepinephrine and serotonin, at the single-vesicle level. The introduction of this methodology marks a significant advancement, offering a novel avenue to explore neurological and psychiatric disorders through the lens of neurotransmitter signaling in the mammalian brain. This breakthrough has the potential to deepen our understanding of the underlying mechanisms of these complex disorders and may pave the way for the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Yuying Liu
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lijiao Cao
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Junlan Zhou
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chuqi Li
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jinchang Du
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yifei Xue
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xianchan Li
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Department of Preventive Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
3
|
Kurogi H, Takasugi N, Kubota S, Kumar A, Suzuki T, Dohmae N, Sawada D, Zhang KY, Uehara T. Discovery of a Compound That Inhibits IRE1α S-Nitrosylation and Preserves the Endoplasmic Reticulum Stress Response under Nitrosative Stress. ACS Chem Biol 2024; 19:2429-2437. [PMID: 39530155 PMCID: PMC11667674 DOI: 10.1021/acschembio.4c00403] [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: 06/10/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Inositol-requiring enzyme 1α (IRE1α) is a sensor of endoplasmic reticulum (ER) stress and drives ER stress response pathways. Activated IRE1α exhibits RNase activity and cleaves mRNA encoding X-box binding protein 1, a transcription factor that induces the expression of genes that maintain ER proteostasis for cell survival. Previously, we showed that IRE1α undergoes S-nitrosylation, a post-translational modification induced by nitric oxide (NO), resulting in reduced RNase activity. Therefore, S-nitrosylation of IRE1α compromises the response to ER stress, making cells more vulnerable. We conducted virtual screening and cell-based validation experiments to identify compounds that inhibit the S-nitrosylation of IRE1α by targeting nitrosylated cysteine residues. We ultimately identified a compound (1ACTA) that selectively inhibits the S-nitrosylation of IRE1α and prevents the NO-induced reduction of RNase activity. Furthermore, 1ACTA reduces the rate of NO-induced cell death. Our research identified S-nitrosylation as a novel target for drug development for IRE1α and provides a suitable screening strategy.
Collapse
Affiliation(s)
- Haruna Kurogi
- Department
of Medicinal Pharmacology, Graduate School of Medicine, Dentistry
and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Nobumasa Takasugi
- Department
of Medicinal Pharmacology, Graduate School of Medicine, Dentistry
and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Sho Kubota
- Department
of Medicinal Pharmacology, Graduate School of Medicine, Dentistry
and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Ashutosh Kumar
- Laboratory
for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Takehiro Suzuki
- Biomolecular
Characterization Unit, Technology Platform Division, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Naoshi Dohmae
- Biomolecular
Characterization Unit, Technology Platform Division, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Daisuke Sawada
- Department
of Fine Organic Synthesis, Graduate School of Medicine, Dentistry
and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Kam Y.J. Zhang
- Laboratory
for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Takashi Uehara
- Department
of Medicinal Pharmacology, Graduate School of Medicine, Dentistry
and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
4
|
Kavrik O, Gumral N, Ozmen O, Aslankoc R, Saygin M, Yalcin A. The combined use of thymoquinone and metformin provides more effective neuroprotection in a mouse model of MPTP-induced Parkinson's disease. J Recept Signal Transduct Res 2024; 44:161-173. [PMID: 39585743 DOI: 10.1080/10799893.2024.2434112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Thymoquinone (TQ) is known for its antioxidant properties, and although metformin (MM) is known as an antidiabetic drug, it is suggested that it reduces neurodegeneration. The study aimed to investigate the neuroprotective effects of TQ and MM, particularly when used together, in relation to Parkinson's disease (PD). In the study, sixty-eight male C57BL/6 mice weighing 25-30 g were divided into five groups as follows: control, MPTP, MPTP+TQ, MPTP+MM, and MPTP+TQ+MM. MM (500 mg/kg, orally) and TQ (5 mg/kg, i.p.) were administered for 21 days. Motor coordination and locomotor activities were evaluated by the pole test. TOS and TAS analyses were conducted to determine oxidative stress levels in the substantia nigra. Dopaminergic degeneration in the substantia nigra was evaluated by analyzing Tyrosine hydroxylase (TH). To evaluate the apoptotic pathway, the expression levels of iNOS, BDNF, Complex 1, Bax, Bcl-2, Cytochrome C, AIF, and Caspase-3 were examined immunohistochemically. Compared to the MPTP-treated group, TQ, MM and MM+TQ treatment provided significant improvement in locomotor activity in mice, significantly increased antioxidant activity, significantly reduced the expression levels of iNOS, Bax, Cytochrome C, Caspase-3, and AIF, significantly increased BDNF, Bcl-2, and Complex 1 expressions, and significantly increased the number of TH positive cells. The separate use of TQ and MM exhibits neuroprotective activity, however, we showed that using TQ and MM in combination may be more effective. This may provide preclinical evidence supporting the therapeutic potential of their combined use for treating PD.
Collapse
Affiliation(s)
- Oguzhan Kavrik
- Department of Physiology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Nurhan Gumral
- Department of Physiology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Ozlem Ozmen
- Department of Pathology, Burdur Mehmet Akif Ersoy University Faculty of Veterinary, Burdur, Turkey
| | - Rahime Aslankoc
- Department of Physiology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Mustafa Saygin
- Department of Physiology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Arzu Yalcin
- Department of Physiology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
5
|
Kartik S, Pal R, Chaudhary MJ, Nath R, Kumar M. Modulation of Autophagy and Nitric Oxide Signaling via Glycyrrhizic Acid and 7-Nitroindazole in MPTP-induced Parkinson's Disease Model. Ann Neurosci 2024; 31:265-276. [PMID: 39840143 PMCID: PMC11744622 DOI: 10.1177/09727531231191661] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/12/2023] [Indexed: 01/23/2025] Open
Abstract
Background Parkinson's disease (PD) is characterized by dopaminergic (DA) neuron loss, Lewy body build-up, and motor dysfunction. One of the primary pathogenic mechanisms of PD development is autophagy dysfunction and nitric oxide-mediated neurotoxicity. Purpose The current study focuses on autophagy and nitric oxide (NO) signaling roles in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated PD mice and their protection by their modulators. Method BALB/c mice were administered MPTP (30 mg/kg/i.p/day) for five consecutive days in order to create a PD model. Following MPTP poisoning, the doses of GA (16.8 mg/kg/day/i.p.), 7-nitroindazole (7-NI) (10 mg/kg/day/i.p.), and their combination were administered once daily for 14 days. Animals were observed for behavioral and locomotor changes, biochemical examination, inflammatory mediators, and analysis of molecular markers. Results GA, 7-NI alone significantly reduced MPTP-induced locomotor, behavioral, and oxidative damage. Additionally, in MPTP-intoxicated animals, 7-NI and GA had protective effects on dopamine levels, TH positive DA neurons, inflammatory cytokines interleukin 1β (IL-1β), tumor necrosis factor-alpha (TNF-α), nuclear factor-kappa B (NF-κB), and cyclooxygenase-2 (Cox-2) concentration. Furthermore, GA increases LC3BII expression, which in turn increases autophagy. It also decreases total NO content, and a significant response of 7-NI demonstrates their interaction, which is neuroprotective. Conclusion Present research suggests that dysregulation of autophagy and NO-mediated neuroinflammation are involved in the pathogenesis and progression of MPTP-induced PD. The use of two pharmacotherapeutics, GA and 7-NI, respectively, significantly reduces MPTP-induced PD distortions and their interaction enhances the overall protective effect, suggesting that these pharmacological agents may be used for the treatment of PD.
Collapse
Affiliation(s)
- Shipra Kartik
- Department of Pharmacology and Therapeutics, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Rishi Pal
- Department of Pharmacology and Therapeutics, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Manju J. Chaudhary
- Department of Physiology, Government Medical College, Tirwa Road, Kannauj, Lucknow, Uttar Pradesh, India
| | - Rajendra Nath
- Department of Pharmacology and Therapeutics, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Madhu Kumar
- Department of Pathology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
6
|
Meng J, Fang J, Bao Y, Chen H, Hu X, Wang Z, Li M, Cheng Q, Dong Y, Yang X, Zou Y, Zhao D, Tang J, Zhang W, Chen C. The biphasic role of Hspb1 on ferroptotic cell death in Parkinson's disease. Theranostics 2024; 14:4643-4666. [PMID: 39239519 PMCID: PMC11373631 DOI: 10.7150/thno.98457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/21/2024] [Indexed: 09/07/2024] Open
Abstract
Rationale: Ferroptosis-driven loss of dopaminergic neurons plays a pivotal role in the pathogenesis of Parkinson's disease (PD). In PD patients, Hspb1 is commonly observed at abnormally high levels in the substantia nigra. The precise consequences of Hspb1 overexpression in PD, however, have yet to be fully elucidated. Methods: We used human iPSC-derived dopaminergic neurons and Coniferaldehyde (CFA)-an Nrf2 agonist known for its ability to cross the blood-brain barrier-to investigate the role of Hspb1 in PD. We examined the correlation between Hspb1 overexpression and Nrf2 activation and explored the transcriptional regulation of Hspb1 by Nrf2. Gene deletion techniques were employed to determine the necessity of Nrf2 and Hspb1 for CFA's neuroprotective effects. Results: Our research demonstrated that Nrf2 can upregulate the transcription of Hspb1 by directly binding to its promoter. Deletion of either Nrf2 or Hspb1 gene abolished the neuroprotective effects of CFA. The Nrf2-Hspb1 pathway, newly identified as a defense mechanism against ferroptosis, was shown to be essential for preventing neurodegeneration progression. Additionally, we discovered that prolonged overexpression of Hspb1 leads to neuronal death and that Hspb1 released from ruptured cells can trigger secondary cell death in neighboring cells, exacerbating neuroinflammatory responses. Conclusions: These findings highlight a biphasic role of Hspb1 in PD, where it initially provides neuroprotection through the Nrf2-Hspb1 pathway but ultimately contributes to neurodegeneration and inflammation when overexpressed. Understanding this dual role is crucial for developing therapeutic strategies targeting Hspb1 and Nrf2 in PD.
Collapse
Affiliation(s)
- Jieyi Meng
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jinyu Fang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yutong Bao
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Huizhu Chen
- School of Clinical Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Xiaodan Hu
- School of Clinical Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Ziyuan Wang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Man Li
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Quancheng Cheng
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yaqiong Dong
- Institute of Translational Medicine, College of Medicine, Qingdao University, Qingdao, Shandong 266023, China
| | - Xiaoda Yang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yushu Zou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Dongyu Zhao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Jiping Tang
- Physiology and Pharmacology Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda 92350, USA
| | - Weiguang Zhang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chunhua Chen
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
7
|
Chauhan P, Pandey P, Khan F, Maqsood R. Insights on the Correlation between Mitochondrial Dysfunction and the Progression of Parkinson's Disease. Endocr Metab Immune Disord Drug Targets 2024; 24:1007-1014. [PMID: 37867265 DOI: 10.2174/0118715303249690231006114308] [Citation(s) in RCA: 1] [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: 02/17/2023] [Revised: 08/09/2023] [Accepted: 08/30/2023] [Indexed: 10/24/2023]
Abstract
The aetiology of a progressive neuronal Parkinson's disease has been discussed in several studies. However, due to the multiple risk factors involved in its development, such as environmental toxicity, parental inheritance, misfolding of protein, ageing, generation of reactive oxygen species, degradation of dopaminergic neurons, formation of neurotoxins, mitochondria dysfunction, and genetic mutations, its mechanism of involvement is still discernible. Therefore, this study aimed to review the processes or systems that are crucially implicated in the conversion of MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) into its lethal form, which directly blockades the performance of mitochondria, leading to the formation of oxidative stress in the dopaminergic neurons of substantia nigra pars compacta (SNpc) and resulting in the progression of an incurable Parkinson's disease. This review also comprises an overview of the mutated genes that are frequently associated with mitochondrial dysfunction and the progression of Parkinson's disease. Altogether, this review would help future researchers to develop an efficient therapeutic approach for the management of Parkinson's disease via identifying potent prognostic and diagnostic biomarkers.
Collapse
Affiliation(s)
- Prashant Chauhan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Noida, India
| | - Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Noida, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Noida, India
| | - Ramish Maqsood
- Department of Biotechnology, Noida Institute of Engineering and Technology, Noida, India
| |
Collapse
|
8
|
Maccallini C, Amoroso R. Neuronal Nitric Oxide Synthase and Post-Translational Modifications in the Development of Central Nervous System Diseases: Implications and Regulation. Molecules 2023; 28:6691. [PMID: 37764469 PMCID: PMC10538099 DOI: 10.3390/molecules28186691] [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: 08/26/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
In the Central Nervous System (CNS), Nitric Oxide (NO) is mainly biosynthesized by neuronal Nitric Oxide Synthase (nNOS). The dysregulated activation of nNOS in neurons is critical in the development of different conditions affecting the CNS. The excessive production of NO by nNOS is responsible for a number of proteins' post-translational modifications (PTMs), which can lead to aberrant biochemical pathways, impairing CNS functions. In this review, we briefly revise the main implications of dysregulated nNOS in the progression of the most prevalent CNS neurodegenerative disorders, i.e., Alzheimer's disease (AD) and Parkinson's disease, as well as in the development of neuronal disorders. Moreover, a specific focus on compounds able to modulate nNOS activity as promising therapeutics to tackle different neuronal diseases is presented.
Collapse
Affiliation(s)
- Cristina Maccallini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy;
| | | |
Collapse
|
9
|
Ozkizilcik A, Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Wiklund L, Sharma HS. Nanowired delivery of antibodies to tau and neuronal nitric oxide synthase together with cerebrolysin attenuates traumatic brain injury induced exacerbation of brain pathology in Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 171:83-121. [PMID: 37783564 DOI: 10.1016/bs.irn.2023.07.001] [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] [Indexed: 10/04/2023]
Abstract
Concussive head injury (CHI) is one of the major risk factors for developing Parkinson's disease in later life of military personnel affecting lifetime functional and cognitive disturbances. Till date no suitable therapies are available to attenuate CHI or PD induced brain pathology. Thus, further exploration of novel therapeutic agents are highly warranted using nanomedicine in enhancing the quality of life of veterans or service members of US military. Since PD or CHI induces oxidative stress and perturbs neurotrophic factors regulation associated with phosphorylated tau (p-tau) deposition, a possibility exists that nanodelivery of agents that could enhance neurotrophic factors balance and attenuate oxidative stress could be neuroprotective in nature. In this review, nanowired delivery of cerebrolysin-a balanced composition of several neurotrophic factors and active peptide fragments together with monoclonal antibodies to neuronal nitric oxide synthase (nNOS) with p-tau antibodies was examined in PD following CHI in model experiments. Our results suggest that combined administration of nanowired antibodies to nNOS and p-tau together with cerebrolysin significantly attenuated CHI induced exacerbation of PD brain pathology. This combined treatment also has beneficial effects in CHI or PD alone, not reported earlier.
Collapse
Affiliation(s)
- Asya Ozkizilcik
- Dept. Biomedical Engineering, University of Arkansas, Fayetteville, AR, United Staes
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
| | - Dafin F Muresanu
- Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; ''RoNeuro'' Institute for Neurological Research and Diagnostic, Mircea Eliade Street, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Department of Anesthesiology, Boston University, Albany str, Boston MA, United States
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
10
|
Bao Y, Gan C, Chen Z, Qi Z, Meng Z, Yue F. Quantification of Non-Motor Symptoms in Parkinsonian Cynomolgus Monkeys. Brain Sci 2023; 13:1153. [PMID: 37626508 PMCID: PMC10452176 DOI: 10.3390/brainsci13081153] [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: 06/01/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder that features motor and non-motor deficits. The use of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopamine neuron degeneration has been widely practiced to produce reliable animal models of PD. However, most previous preclinical studies focused on motor dysfunction, and few non-motor symptoms were evaluated. Thus far, there is a lack of comprehensive investigations of the non-motor symptoms in animal models. OBJECTIVES In this study, we aim to use a battery of behavioral methods to evaluate non-motor symptoms in MPTP-induced non-human primate PD models. METHODS Cognitive function, sleep, and psychiatric behaviors were evaluated in MPTP-treated cynomolgus monkeys. The tests consisted of a delayed matching-to-sample (DMTS) task, the use of a physical activity monitor (PAM), an apathy feeding task (AFT), the human intruder test (HIT), novel fruit test (NFT), and predator confrontation test (PCT). In addition, we tested whether the dopamine receptor agonist pramipexole (PPX) can improve these non-motor symptoms. RESULTS The present results show that the MPTP-treated monkeys exhibited cognitive deficits, abnormal sleep, and anxiety-like behaviors when compared to the control monkeys. These symptoms were relieved partially by PPX. CONCLUSIONS These results suggest that MPTP-induced PD monkeys displayed non-motor symptoms that were similar to those found in PD patients. PPX treatment showed moderate therapeutic effects on these non-motor symptoms. This battery of behavioral tests may provide a valuable model for future preclinical research.
Collapse
Affiliation(s)
- Yu Bao
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.B.)
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chaoning Gan
- Medical College of Guangxi University, Nanning 530003, China
| | - Zuyue Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.B.)
| | - Zhongquan Qi
- Medical College of Guangxi University, Nanning 530003, China
| | - Zhiqiang Meng
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.B.)
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Feng Yue
- Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| |
Collapse
|
11
|
Iova OM, Marin GE, Lazar I, Stanescu I, Dogaru G, Nicula CA, Bulboacă AE. Nitric Oxide/Nitric Oxide Synthase System in the Pathogenesis of Neurodegenerative Disorders-An Overview. Antioxidants (Basel) 2023; 12:antiox12030753. [PMID: 36979000 PMCID: PMC10045816 DOI: 10.3390/antiox12030753] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/24/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Nitric oxide, a ubiquitous molecule found throughout the natural world, is a key molecule implicated in many central and benefic molecular pathways and has a well-established role in the function of the central nervous system, as numerous studies have previously shown. Dysregulation of its metabolism, mainly the upregulation of nitric oxide production, has been proposed as a trigger and/or aggravator for many neurological affections. Increasing evidence supports the implication of this molecule in prevalent neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, or amyotrophic lateral sclerosis. The mechanisms proposed for its neurotoxicity mainly center around the increased quantities of nitric oxide that are produced in the brain, their cause, and, most importantly, the pathological metabolic cascades created. These cascades lead to the formation of neuronal toxic substances that impair the neurons' function and structure on multiple levels. The purpose of this review is to present the main causes of increased pathological production, as well as the most important pathophysiological mechanisms triggered by nitric oxide, mechanisms that could help explain a part of the complex picture of neurodegenerative diseases and help develop targeted therapies.
Collapse
Affiliation(s)
- Olga-Maria Iova
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Gheorghe-Eduard Marin
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Izabella Lazar
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Stanescu
- Department of Neurology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Gabriela Dogaru
- Department of Physical Medicine and Rehabilitation, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Viilor Street, No. 46-50, 400347 Cluj-Napoca, Romania
| | - Cristina Ariadna Nicula
- Department of Ophthalmology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Adriana Elena Bulboacă
- Department of Pathophysiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
12
|
Steinert JR, Amal H. The contribution of an imbalanced redox signalling to neurological and neurodegenerative conditions. Free Radic Biol Med 2023; 194:71-83. [PMID: 36435368 DOI: 10.1016/j.freeradbiomed.2022.11.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Nitric oxide and other redox active molecules such as oxygen free radicals provide essential signalling in diverse neuronal functions, but their excess production and insufficient scavenging induces cytotoxic redox stress which is associated with numerous neurodegenerative and neurological conditions. A further component of redox signalling is mediated by a homeostatic regulation of divalent metal ions, the imbalance of which contributes to neuronal dysfunction. Additional antioxidant molecules such as glutathione and enzymes such as super oxide dismutase are involved in maintaining a physiological redox status within neurons. When cellular processes are perturbed and generation of free radicals overwhelms the antioxidants capacity of the neurons, a resulting redox damage leads to neuronal dysfunction and cell death. Cellular sources for production of redox-active molecules may include NADPH oxidases, mitochondria, cytochrome P450 and nitric oxide (NO)-generating enzymes, such as endothelial, neuronal and inducible NO synthases. Several neurodegenerative and developmental neurological conditions are associated with an imbalanced redox state as a result of neuroinflammatory processes leading to nitrosative and oxidative stress. Ongoing research aims at understanding the causes and consequences of such imbalanced redox homeostasis and its role in neuronal dysfunction.
Collapse
Affiliation(s)
- Joern R Steinert
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham, NG7 2NR, UK.
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
13
|
Yang X, Li L, Shi Y, Wang X, Zhang Y, Jin M, Chen X, Wang R, Liu K. Neurotoxicity of sanguinarine via inhibiting mitophagy and activating apoptosis in zebrafish and PC12 cells. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2022; 188:105259. [PMID: 36464364 DOI: 10.1016/j.pestbp.2022.105259] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/20/2022] [Accepted: 10/06/2022] [Indexed: 06/17/2023]
Abstract
Sanguinarine, a plant-derived phytoalexin, displays various biological activities, such as insecticidal, antimicrobial, anti-inflammatory, anti-angiogenesis and antitumor effects. But its potential neurotoxicity and the underlying mechanisms has rarely been investigated. Therefore, we aimed to assess the neurotoxicity of sanguinarine using zebrafish model and PC12 cells in this study. The results showed that sanguinarine induced the reduction of the length of dopamine neurons and inhibited the blood vessel in the head area of the zebrafish. Further studies demonstrated that the behavioral phenotype of the larval zebrafish was changed by sanguinarine. In addition, there were more apoptotic cells in the larval zebrafish head area. The mRNA expression levels of β-syn, th, pink1 and parkin, closely related to the nervous function, were changed after sanguinarine treatment. The in vitro studies show that notably increases of ROS and apoptosis levels in PC12 cells were observed after sanguinarine treatment. Moreover, the protein expression of Caspase3, Parp, Bax, Bcl2, α-Syn, Th, PINK1 and Parkin were also altered by sanguinarine. Our data indicated that the inhibition of mitophagy, ROS elevation and apoptosis were involved in the neurotoxicity of sanguinarine. These findings will be useful to understand the toxicity induced by sanguinarine.
Collapse
Affiliation(s)
- Xueliang Yang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Lei Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Yuxin Shi
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Xue Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Xiqiang Chen
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| |
Collapse
|
14
|
Martins-Pinge MC, de Jager L, de Campos BH, Bezerra LO, Turini PG, Pinge-Filho P. Nitric Oxide Involvement in Cardiovascular Dysfunctions of Parkinson Disease. Front Pharmacol 2022; 13:898797. [PMID: 35899105 PMCID: PMC9309809 DOI: 10.3389/fphar.2022.898797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra, causing motor changes. In addition to motor symptoms, non-motor dysfunctions such as psychological, sensory and autonomic disorders are recorded. Manifestations related to the autonomic nervous system include the cardiovascular system, as postural hypotension, postprandial hypotension, and low blood pressure. One of the mediators involved is the nitric oxide (NO). In addition to the known roles such as vasodilator, neuromodulator, NO acts as an important mediator of the immune response, increasing the inflammatory response provoked by PD in central nervous system. The use of non-specific NOS inhibitors attenuated the neurodegenerative response in animal models of PD. However, the mechanisms by which NO contributes to neurodegeneration are still not well understood. The literature suggest that the contribution of NO occurs through its interaction with superoxides, products of oxidative stress, and blocking of the mitochondrial respiratory chain, resulting in neuronal death. Most studies involving Parkinsonism models have evaluated brain NO concentrations, with little data available on its peripheral action. Considering that studies that evaluated the involvement of NO in the neurodegeneration in PD, through NOS inhibitors administration, showed neuroprotection in rats, it has prompted new studies to assess the participation of NOS isoforms in cardiovascular changes induced by parkinsonism, and thus to envision new targets for the treatment of cardiovascular disorders in PD. The aim of this study was to conduct a literature review to assess available information on the involvement of nitric oxide (NO) in cardiovascular aspects of PD.
Collapse
Affiliation(s)
- Marli Cardoso Martins-Pinge
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina—UEL, Londrina, Brazil
- *Correspondence: Marli Cardoso Martins-Pinge,
| | - Lorena de Jager
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina—UEL, Londrina, Brazil
| | - Blenda Hyedra de Campos
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina—UEL, Londrina, Brazil
| | - Lorena Oliveira Bezerra
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina—UEL, Londrina, Brazil
| | - Pamela Giovana Turini
- Departamento de Ciências Fisiológicas, Universidade Estadual de Londrina—UEL, Londrina, Brazil
| | - Phileno Pinge-Filho
- Departamento de Ciências Patológicas, Universidade Estadual de Londrina- UEL, Londrina, Brazil
| |
Collapse
|
15
|
Goiran T, Eldeeb MA, Zorca CE, Fon EA. Hallmarks and Molecular Tools for the Study of Mitophagy in Parkinson’s Disease. Cells 2022; 11:cells11132097. [PMID: 35805181 PMCID: PMC9265644 DOI: 10.3390/cells11132097] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/27/2023] Open
Abstract
The best-known hallmarks of Parkinson’s disease (PD) are the motor deficits that result from the degeneration of dopaminergic neurons in the substantia nigra. Dopaminergic neurons are thought to be particularly susceptible to mitochondrial dysfunction. As such, for their survival, they rely on the elaborate quality control mechanisms that have evolved in mammalian cells to monitor mitochondrial function and eliminate dysfunctional mitochondria. Mitophagy is a specialized type of autophagy that mediates the selective removal of damaged mitochondria from cells, with the net effect of dampening the toxicity arising from these dysfunctional organelles. Despite an increasing understanding of the molecular mechanisms that regulate the removal of damaged mitochondria, the detailed molecular link to PD pathophysiology is still not entirely clear. Herein, we review the fundamental molecular pathways involved in PINK1/Parkin-mediated and receptor-mediated mitophagy, the evidence for the dysfunction of these pathways in PD, and recently-developed state-of-the art assays for measuring mitophagy in vitro and in vivo.
Collapse
|
16
|
Lim HS, Park G. Resilin, an insect-derived elastomeric protein, protects dopaminergic neurons in Parkinson disease models. Neurosci Lett 2022; 781:136667. [DOI: 10.1016/j.neulet.2022.136667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
|
17
|
Park H, Kam TI, Peng H, Chou SC, Mehrabani-Tabari AA, Song JJ, Yin X, Karuppagounder SS, Umanah GK, Rao AVS, Choi Y, Aggarwal A, Chang S, Kim H, Byun J, Liu JO, Dawson TM, Dawson VL. PAAN/MIF nuclease inhibition prevents neurodegeneration in Parkinson's disease. Cell 2022; 185:1943-1959.e21. [PMID: 35545089 DOI: 10.1016/j.cell.2022.04.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/14/2022] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
Parthanatos-associated apoptosis-inducing factor (AIF) nuclease (PAAN), also known as macrophage migration inhibitor factor (MIF), is a member of the PD-D/E(X)K nucleases that acts as a final executioner in parthanatos. PAAN's role in Parkinson's disease (PD) and whether it is amenable to chemical inhibition is not known. Here, we show that neurodegeneration induced by pathologic α-synuclein (α-syn) occurs via PAAN/MIF nuclease activity. Genetic depletion of PAAN/MIF and a mutant lacking nuclease activity prevent the loss of dopaminergic neurons and behavioral deficits in the α-syn preformed fibril (PFF) mouse model of sporadic PD. Compound screening led to the identification of PAANIB-1, a brain-penetrant PAAN/MIF nuclease inhibitor that prevents neurodegeneration induced by α-syn PFF, AAV-α-syn overexpression, or MPTP intoxication in vivo. Our findings could have broad relevance in human pathologies where parthanatos plays a role in the development of cell death inhibitors targeting the druggable PAAN/MIF nuclease.
Collapse
Affiliation(s)
- Hyejin Park
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Tae-In Kam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Hanjing Peng
- Department of Pharmacology and Molecular Sciences and SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shih-Ching Chou
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amir A Mehrabani-Tabari
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jae-Jin Song
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xiling Yin
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Senthilkumar S Karuppagounder
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - George K Umanah
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - A V Subba Rao
- Department of Pharmacology and Molecular Sciences and SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - YuRee Choi
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Akanksha Aggarwal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sohyun Chang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hyunhee Kim
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jiyoung Byun
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences and SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
18
|
6-Benzyloxyphthalides as selective and reversible monoamine oxidase B inhibitors with antioxidant and anti-neuroinflammatory activities for Parkinson’s disease treatment. Bioorg Chem 2022; 120:105623. [DOI: 10.1016/j.bioorg.2022.105623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/13/2021] [Accepted: 01/11/2022] [Indexed: 01/05/2023]
|
19
|
Bashirzade AAO, Cheresiz SV, Belova AS, Drobkov AV, Korotaeva AD, Azizi-Arani S, Azimirad A, Odle E, Gild EYV, Ardashov OV, Volcho KP, Bozhko DV, Myrov VO, Kolchanova SM, Polovian AI, Galumov GK, Salakhutdinov NF, Amstislavskaya TG, Kalueff AV. MPTP-Treated Zebrafish Recapitulate 'Late-Stage' Parkinson's-like Cognitive Decline. TOXICS 2022; 10:69. [PMID: 35202255 PMCID: PMC8879925 DOI: 10.3390/toxics10020069] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 11/25/2022]
Abstract
The zebrafish is a promising model species in biomedical research, including neurotoxicology and neuroactive drug screening. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) evokes degeneration of dopaminergic neurons and is commonly used to model Parkinson's disease (PD) in laboratory animals, including zebrafish. However, cognitive phenotypes in MPTP-evoked experimental PD models remain poorly understood. Here, we established an LD50 (292 mg/kg) for intraperitoneal MPTP administration in adult zebrafish, and report impaired spatial working memory (poorer spontaneous alternation in the Y-maze) in a PD model utilizing fish treated with 200 µg of this agent. In addition to conventional behavioral analyses, we also employed artificial intelligence (AI)-based approaches to independently and without bias characterize MPTP effects on zebrafish behavior during the Y-maze test. These analyses yielded a distinct cluster for 200-μg MPTP (vs. other) groups, suggesting that high-dose MPTP produced distinct, computationally detectable patterns of zebrafish swimming. Collectively, these findings support MPTP treatment in adult zebrafish as a late-stage experimental PD model with overt cognitive phenotypes.
Collapse
Affiliation(s)
- Alim A. O. Bashirzade
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Sergey V. Cheresiz
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Alisa S. Belova
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Alexey V. Drobkov
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Anastasiia D. Korotaeva
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Soheil Azizi-Arani
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Amirhossein Azimirad
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Eric Odle
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Emma-Yanina V. Gild
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Oleg V. Ardashov
- Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, 630090 Novosibirsk, Russia; (O.V.A.); (K.P.V.); (N.F.S.)
| | - Konstantin P. Volcho
- Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, 630090 Novosibirsk, Russia; (O.V.A.); (K.P.V.); (N.F.S.)
| | - Dmitrii V. Bozhko
- ZebraML, Inc., Houston, TX 77043, USA; (D.V.B.); (V.O.M.); (S.M.K.); (A.I.P.); (G.K.G.)
| | - Vladislav O. Myrov
- ZebraML, Inc., Houston, TX 77043, USA; (D.V.B.); (V.O.M.); (S.M.K.); (A.I.P.); (G.K.G.)
| | - Sofia M. Kolchanova
- ZebraML, Inc., Houston, TX 77043, USA; (D.V.B.); (V.O.M.); (S.M.K.); (A.I.P.); (G.K.G.)
| | | | - Georgii K. Galumov
- ZebraML, Inc., Houston, TX 77043, USA; (D.V.B.); (V.O.M.); (S.M.K.); (A.I.P.); (G.K.G.)
| | - Nariman F. Salakhutdinov
- Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, 630090 Novosibirsk, Russia; (O.V.A.); (K.P.V.); (N.F.S.)
| | - Tamara G. Amstislavskaya
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Allan V. Kalueff
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
- Ural Federal University, 620002 Yekaterinburg, Russia
- Neurobiology Program, Sirius University of Science and Technology, 354340 Sochi, Russia
- Moscow Institute of Physics and Technology, 141701 Moscow, Russia
- Granov Scientific Research Center of Radiology and Surgical Technologies, 197758 St. Petersburg, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia
- School of Pharmacy, Southwest University, Chongqing 400715, China
| |
Collapse
|
20
|
He D, Hu G, Zhou A, Liu Y, Huang B, Su Y, Wang H, Ye B, He Y, Gao X, Fu S, Liu D. Echinocystic Acid Inhibits Inflammation and Exerts Neuroprotective Effects in MPTP-Induced Parkinson’s Disease Model Mice. Front Pharmacol 2022; 12:787771. [PMID: 35126128 PMCID: PMC8807489 DOI: 10.3389/fphar.2021.787771] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/29/2021] [Indexed: 01/05/2023] Open
Abstract
Parkinson’s disease (PD), the second primary neurodegenerative disease affecting human health, is mainly characterized by dopaminergic neuron damage in the midbrain and the clinical manifestation of movement disorders. Studies have shown that neuroinflammation plays an important role in the progression of PD. Excessively activated microglia produce several pro-inflammatory mediators, leading to damage to the surrounding neurons and finally inducing neurodegeneration. Echinocystic acid (EA) exhibits an anti-inflammatory effect in peripheral tissues. However, whether it inhibited neuroinflammation remains unclear. Therefore, the current study investigates the effect of EA on neuroinflammation and whether it can improve PD symptoms through inhibiting neuroinflammation. In our experiments, we discovered that EA inhibited the production of pro-inflammatory mediators in LPS-exposed BV2 cells. Further mechanism-related studies revealed that EA inhibited inflammation by activating PI3K/Akt and inhibiting NF-κB and MAPK signal pathways in LPS-induced BV2 cells. Research revealed that EA eases microglia-mediated neuron death in SN4741 and SHSY5Y cells. In in vivo studies, the results demonstrated that EA improves weight loss and behavioral impairment in MPTP-induced mice. Further studies have revealed that EA inhibited dopaminergic neuron damage and inflammation in the mice midbrain. In conclusion, our study demonstrated that EA inhibits neuroinflammation and exerts neuroprotective effects by activating PI3K/Akt and inhibiting NF-κB and MAPK signal pathways in vivo and in vitro.
Collapse
Affiliation(s)
- Dewei He
- College of Animal Science, Jilin University, Changchun, China
| | - Guiqiu Hu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ang Zhou
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yanting Liu
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Bingxu Huang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yingchun Su
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hefei Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Bojian Ye
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuan He
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiyu Gao
- College of Animal Science, Jilin University, Changchun, China
| | - Shoupeng Fu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Dianfeng Liu
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
21
|
Mechanistic Insights Expatiating the Redox-Active-Metal-Mediated Neuronal Degeneration in Parkinson's Disease. Int J Mol Sci 2022; 23:ijms23020678. [PMID: 35054862 PMCID: PMC8776156 DOI: 10.3390/ijms23020678] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is a complicated and incapacitating neurodegenerative malady that emanates following the dopaminergic (DArgic) nerve cell deprivation in the substantia nigra pars compacta (SN-PC). The etiopathogenesis of PD is still abstruse. Howbeit, PD is hypothesized to be precipitated by an amalgamation of genetic mutations and exposure to environmental toxins. The aggregation of α-synucelin within the Lewy bodies (LBs), escalated oxidative stress (OS), autophagy-lysosome system impairment, ubiquitin-proteasome system (UPS) impairment, mitochondrial abnormality, programmed cell death, and neuroinflammation are regarded as imperative events that actively participate in PD pathogenesis. The central nervous system (CNS) relies heavily on redox-active metals, particularly iron (Fe) and copper (Cu), in order to modulate pivotal operations, for instance, myelin generation, synthesis of neurotransmitters, synaptic signaling, and conveyance of oxygen (O2). The duo, namely, Fe and Cu, following their inordinate exposure, are viable of permeating across the blood–brain barrier (BBB) and moving inside the brain, thereby culminating in the escalated OS (through a reactive oxygen species (ROS)-reliant pathway), α-synuclein aggregation within the LBs, and lipid peroxidation, which consequently results in the destruction of DArgic nerve cells and facilitates PD emanation. This review delineates the metabolism of Fe and Cu in the CNS, their role and disrupted balance in PD. An in-depth investigation was carried out by utilizing the existing publications obtained from prestigious medical databases employing particular keywords mentioned in the current paper. Moreover, we also focus on decoding the role of metal complexes and chelators in PD treatment. Conclusively, metal chelators hold the aptitude to elicit the scavenging of mobile/fluctuating metal ions, which in turn culminates in the suppression of ROS generation, and thereby prelude the evolution of PD.
Collapse
|
22
|
Kalous KS, Wynia-Smith SL, Smith BC. Sirtuin Oxidative Post-translational Modifications. Front Physiol 2021; 12:763417. [PMID: 34899389 PMCID: PMC8652059 DOI: 10.3389/fphys.2021.763417] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Increased sirtuin deacylase activity is correlated with increased lifespan and healthspan in eukaryotes. Conversely, decreased sirtuin deacylase activity is correlated with increased susceptibility to aging-related diseases. However, the mechanisms leading to decreased sirtuin activity during aging are poorly understood. Recent work has shown that oxidative post-translational modification by reactive oxygen (ROS) or nitrogen (RNS) species results in inhibition of sirtuin deacylase activity through cysteine nitrosation, glutathionylation, sulfenylation, and sulfhydration as well as tyrosine nitration. The prevalence of ROS/RNS (e.g., nitric oxide, S-nitrosoglutathione, hydrogen peroxide, oxidized glutathione, and peroxynitrite) is increased during inflammation and as a result of electron transport chain dysfunction. With age, cellular production of ROS/RNS increases; thus, cellular oxidants may serve as a causal link between loss of sirtuin activity and aging-related disease development. Therefore, the prevention of inhibitory oxidative modification may represent a novel means to increase sirtuin activity during aging. In this review, we explore the role of cellular oxidants in inhibiting individual sirtuin human isoform deacylase activity and clarify the relevance of ROS/RNS as regulatory molecules of sirtuin deacylase activity in the context of health and disease.
Collapse
Affiliation(s)
- Kelsey S Kalous
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sarah L Wynia-Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
23
|
Cao Y, Li B, Ismail N, Smith K, Li T, Dai R, Deng Y. Neurotoxicity and Underlying Mechanisms of Endogenous Neurotoxins. Int J Mol Sci 2021; 22:12805. [PMID: 34884606 PMCID: PMC8657695 DOI: 10.3390/ijms222312805] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/16/2022] Open
Abstract
Endogenous and exogenous neurotoxins are important factors leading to neurodegenerative diseases. In the 1980s, the discovery that 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) contributes to Parkinson's disease (PD) symptoms led to new research investigations on neurotoxins. An abnormal metabolism of endogenous substances, such as condensation of bioamines with endogenous aldehydes, dopamine (DA) oxidation, and kynurenine pathway, can produce endogenous neurotoxins. Neurotoxins may damage the nervous system by inhibiting mitochondrial activity, increasing oxidative stress, increasing neuroinflammation, and up-regulating proteins related to cell death. This paper reviews the biological synthesis of various known endogenous neurotoxins and their toxic mechanisms.
Collapse
Affiliation(s)
- Yanlu Cao
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Y.C.); (T.L.); (Y.D.)
| | - Bo Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Y.C.); (T.L.); (Y.D.)
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Nafissa Ismail
- Neuroimmunology, Stress and Endocrinology (NISE) Lab, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (N.I.); (K.S.)
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Kevin Smith
- Neuroimmunology, Stress and Endocrinology (NISE) Lab, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (N.I.); (K.S.)
| | - Tianmei Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Y.C.); (T.L.); (Y.D.)
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
| | - Rongji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Y.C.); (T.L.); (Y.D.)
| | - Yulin Deng
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Y.C.); (T.L.); (Y.D.)
| |
Collapse
|
24
|
Pirooznia SK, Rosenthal LS, Dawson VL, Dawson TM. Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection. Pharmacol Rev 2021; 73:33-97. [PMID: 34663684 DOI: 10.1124/pharmrev.120.000189] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheila K Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Liana S Rosenthal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| |
Collapse
|
25
|
Srivastava R, Choudhury PK, Dev SK, Rathore V. Neuroprotective effect of α-pinene self-emulsifying nanoformulation against 6-OHDA induced neurotoxicity on human SH-SY5Y cells and its in vivo validation for anti-Parkinson's effect. J Biochem Mol Toxicol 2021; 35:e22902. [PMID: 34464010 DOI: 10.1002/jbt.22902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 08/15/2021] [Accepted: 08/20/2021] [Indexed: 11/07/2022]
Abstract
Oxidative stress (OS) is involved in the multifaceted pathogenic paradigm of neurodegenerative diseases like Parkinson's disease (PD). Monoterpenes like α-pinene (ALP) is considered to be a therapeutically potent antioxidant agent able to attenuate and scavenge various reactive oxygen species and reactive nitrogen species. The present study aimed to evaluate the in vitro and in vivo neuroprotective effect of α-pinene self-emulsifying nanoformulation (ALP-SENF) for PD. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay was done to evaluate the neurotoxic dose of the ALP-SENF; however, the neuroprotective effect was assessed by 6-hydroxydopamine (6-OHDA) induced neurotoxicity model on SH-SY5Y taking NAC (N-acetyl-l-cysteine) as standard. The in vivo anti-Parkinson's activity of the ALP-SENF was compared with that of the plain ALP suspension by using reserpine antagonism and haloperidol-induced Parkinsonism model in rats. Various behavioral tests and biochemical antioxidant enzymes were estimated. The in vitro results revealed that treatment with ALP-SENF at a concentration of 100 and 200 µM was found to show significant neuronal SH-SY5Y cell viability against 50 µM 6-OHDA. ALP-SENF treated animals have seen significant neurobehavioral improvement. Furthermore, the levels of antioxidative enzymes in biochemical test reveals a marked enhancement in the expression of antioxidant enzymes that significantly attenuated the OS induced neurodegeneration. Due to the mechanisms of their antioxidant action, it was probably due to the scavenging of free radicals and the expression of antioxidant enzymes. It also improved neurobehavioral changes induced by reserpine and haloperidol.
Collapse
Affiliation(s)
- Rajnish Srivastava
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, India
| | - Pratim K Choudhury
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, India
| | - Suresh K Dev
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, India
| | - Vaibhav Rathore
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, India
| |
Collapse
|
26
|
Cansız D, Ustundag UV, Unal I, Alturfan AA, Emekli-Alturfan E. Morphine attenuates neurotoxic effects of MPTP in zebrafish embryos by regulating oxidant/antioxidant balance and acetylcholinesterase activity. Drug Chem Toxicol 2021; 45:2439-2447. [PMID: 34340603 DOI: 10.1080/01480545.2021.1957558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases due to the loss of dopaminergic neurons in the midbrain in the substantia nigra. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a neurotoxic agent causing disruptions in mitochondria of dopaminergic neurons leading to impaired oxidant-antioxidant balance. Both zebrafish and zebrafish embryos are sensitive to MPTP. In zebrafish embryos, MPTP decreases the dopaminergic cells in the diencephalon by damaging dopaminergic neurons. Morphine is an opioid pain killer and a strong analgesic that is used to treat chronic pain. Until today morphine has been shown to regulate the survival or death of neurons and both protective and destructive effects of morphine have been reported in the central nervous system. This study aimed to evaluate the effects of morphine in MPTP-exposed zebrafish embryos. Developmental parameters were monitored and documented daily during embryonic development. Locomotor activity of zebrafish embryos at 96 h postfertilization (hpf) was determined. Acetylcholinesterase (AChE) activity and oxidant-antioxidant parameters were analyzed by biochemical methods. RT-PCR was used to evaluate bdnf, dj1, lrrk and pink1 expressions. Morphine treatment improved mortality and hatching rates, locomotor activity, AChE, and antioxidant enzyme activities as well as the expressions of bdnf, dj1, lrrk and pink1 in a dose-dependent manner that were altered by MPTP. Increased lipid peroxidation supports the role of morphine to induce autophagy to prevent PD-related pathologies. Our study provided important data on the possible molecular mechanism of the therapeutic effects of morphine in PD.
Collapse
Affiliation(s)
- Derya Cansız
- Faculty of Medicine, Department of Biochemistry, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Unsal Veli Ustundag
- Faculty of Medicine, Department of Medical Biochemistry, Istanbul Medipol University, Istanbul, Turkey
| | - Ismail Unal
- Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - A Ata Alturfan
- Faculty of Medicine, Department of Biochemistry, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Ebru Emekli-Alturfan
- Faculty of Dentistry, Department of Basic Medical Sciences, Marmara University, Istanbul, Turkey
| |
Collapse
|
27
|
Ma L, Gholam Azad M, Dharmasivam M, Richardson V, Quinn RJ, Feng Y, Pountney DL, Tonissen KF, Mellick GD, Yanatori I, Richardson DR. Parkinson's disease: Alterations in iron and redox biology as a key to unlock therapeutic strategies. Redox Biol 2021; 41:101896. [PMID: 33799121 PMCID: PMC8044696 DOI: 10.1016/j.redox.2021.101896] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
A plethora of studies indicate that iron metabolism is dysregulated in Parkinson's disease (PD). The literature reveals well-documented alterations consistent with established dogma, but also intriguing paradoxical observations requiring mechanistic dissection. An important fact is the iron loading in dopaminergic neurons of the substantia nigra pars compacta (SNpc), which are the cells primarily affected in PD. Assessment of these changes reveal increased expression of proteins critical for iron uptake, namely transferrin receptor 1 and the divalent metal transporter 1 (DMT1), and decreased expression of the iron exporter, ferroportin-1 (FPN1). Consistent with this is the activation of iron regulator protein (IRP) RNA-binding activity, which is an important regulator of iron homeostasis, with its activation indicating cytosolic iron deficiency. In fact, IRPs bind to iron-responsive elements (IREs) in the 3ꞌ untranslated region (UTR) of certain mRNAs to stabilize their half-life, while binding to the 5ꞌ UTR prevents translation. Iron loading of dopaminergic neurons in PD may occur through these mechanisms, leading to increased neuronal iron and iron-mediated reactive oxygen species (ROS) generation. The "gold standard" histological marker of PD, Lewy bodies, are mainly composed of α-synuclein, the expression of which is markedly increased in PD. Of note, an atypical IRE exists in the α-synuclein 5ꞌ UTR that may explain its up-regulation by increased iron. This dysregulation could be impacted by the unique autonomous pacemaking of dopaminergic neurons of the SNpc that engages L-type Ca+2 channels, which imparts a bioenergetic energy deficit and mitochondrial redox stress. This dysfunction could then drive alterations in iron trafficking that attempt to rescue energy deficits such as the increased iron uptake to provide iron for key electron transport proteins. Considering the increased iron-loading in PD brains, therapies utilizing limited iron chelation have shown success. Greater therapeutic advancements should be possible once the exact molecular pathways of iron processing are dissected.
Collapse
Affiliation(s)
- L Ma
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Gholam Azad
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Dharmasivam
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - V Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - R J Quinn
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - Y Feng
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - D L Pountney
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - K F Tonissen
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - G D Mellick
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - I Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - D R Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
28
|
Wang X, Ge P. Parthanatos in the pathogenesis of nervous system diseases. Neuroscience 2020; 449:241-250. [DOI: 10.1016/j.neuroscience.2020.09.049] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
|
29
|
Coimbra DF, Cintra CH, Lourenço LCL, Parreira RLT, Orenha RP, Caramori GF. Are DFT Methods Able to Predict Reduction Potentials of Ruthenium Nitrosyl Complexes Accurately? J Phys Chem A 2020; 124:6186-6192. [DOI: 10.1021/acs.jpca.0c03718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Daniel F. Coimbra
- Departamento de Química, Universidade Federal de Santa Catarina, Campus Universitário Trindade, CP 476, Florianópolis, SC 88040−900, Brazil
| | - Claudia H. Cintra
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Franca, SP, 14404-600 Brazil
| | - Luiz C. L. Lourenço
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Franca, SP, 14404-600 Brazil
| | - Renato L. T. Parreira
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Franca, SP, 14404-600 Brazil
| | - Renato P. Orenha
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Franca, SP, 14404-600 Brazil
| | - Giovanni F. Caramori
- Departamento de Química, Universidade Federal de Santa Catarina, Campus Universitário Trindade, CP 476, Florianópolis, SC 88040−900, Brazil
| |
Collapse
|
30
|
S-Nitrosylation of G protein-coupled receptor kinase 6 and Casein kinase 2 alpha modulates their kinase activity toward alpha-synuclein phosphorylation in an animal model of Parkinson's disease. PLoS One 2020; 15:e0232019. [PMID: 32343709 PMCID: PMC7188290 DOI: 10.1371/journal.pone.0232019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 04/06/2020] [Indexed: 01/02/2023] Open
Abstract
Parkinson’s disease (PD) is a common neurodegenerative disorder which is mostly sporadic but familial-linked PD (FPD) cases have also been found. The first reported gene mutation that linked to PD is α-synuclein (α-syn). Studies have shown that mutations, increased expression or abnormal processing of α-syn can contribute to PD, but it is believed that multiple mechanisms are involved. One of the contributing factors is post-translational modification (PTM), such as phosphorylation of α-syn at serine 129 by G-protein-coupled receptor kinases (GRKs) and casein kinase 2α (CK2α). Another known important contributing factor to PD pathogenesis is oxidative and nitrosative stress. In this study, we found that GRK6 and CK2α can be S-nitrosylated by nitric oxide (NO) both in vitro and in vivo. S-nitrosylation of GRK6 and CK2α enhanced their kinase activity towards the phosphorylation of α-syn at S129. In an A53T α-syn transgenic mouse model of PD, we found that increased GRK6 and CK2α S-nitrosylation were observed in an age dependent manner and it was associated with an increased level of pSer129 α-syn. Treatment of A53T α-syn transgenic mice with Nω-Nitro-L-arginine (L-NNA) significantly reduced the S-nitrosylation of GRK6 and CK2α in the brain. Finally, deletion of neuronal nitric oxide synthase (nNOS) in A53T α-syn transgenic mice reduced the levels of pSer129 α-syn and α-syn in an age dependent manner. Our results provide a novel mechanism of how NO through S-nitrosylation of GRK6 and CK2α can enhance the phosphorylation of pSer129 α-syn in an animal model of PD.
Collapse
|
31
|
Nakamura T, Lipton SA. Nitric Oxide-Dependent Protein Post-Translational Modifications Impair Mitochondrial Function and Metabolism to Contribute to Neurodegenerative Diseases. Antioxid Redox Signal 2020; 32:817-833. [PMID: 31657228 PMCID: PMC7074890 DOI: 10.1089/ars.2019.7916] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Significance: Most brains affected by neurodegenerative diseases manifest mitochondrial dysfunction as well as elevated production of reactive oxygen species and reactive nitrogen species (RNS), contributing to synapse loss and neuronal injury. Recent Advances: Excessive production of RNS triggers nitric oxide (NO)-mediated post-translational modifications of proteins, such as S-nitrosylation of cysteine residues and nitration of tyrosine residues. Proteins thus affected impair mitochondrial metabolism, mitochondrial dynamics, and mitophagy in the nervous system. Critical Issues: Identification and better characterization of underlying molecular mechanisms for NO-mediated mitochondrial dysfunction will provide important insights into the pathogenesis of neurodegenerative disorders. In this review, we highlight recent discoveries concerning S-nitrosylation of the tricarboxylic acid cycle enzymes, mitochondrial fission GTPase dynamin-related protein 1, and mitophagy-related proteins Parkin and phosphatase and tensin homolog-induced putative kinase protein 1. We delineate signaling cascades affected by pathologically S-nitrosylated proteins that diminish mitochondrial function in neurodegenerative diseases. Future Directions: Further elucidation of the pathological events resulting from aberrant S-nitrosothiol or nitrotyrosine formation may lead to new therapeutic approaches to ameliorate neurodegenerative disorders.
Collapse
Affiliation(s)
- Tomohiro Nakamura
- Departments of Molecular Medicine and Neuroscience, Neuroscience Translational Center, The Scripps Research Institute, La Jolla, California
- Address correspondence to: Dr. Tomohiro Nakamura, Departments of Molecular Medicine and Neuroscience, Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037
| | - Stuart A. Lipton
- Departments of Molecular Medicine and Neuroscience, Neuroscience Translational Center, The Scripps Research Institute, La Jolla, California
- Department of Neurosciences, University of California San Diego, School of Medicine, La Jolla, California
- Dr. Stuart A. Lipton, Departments of Molecular Medicine and Neuroscience, Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
32
|
Franco R, Rivas-Santisteban R, Reyes-Resina I, Navarro G, Martínez-Pinilla E. Microbiota and Other Preventive Strategies and Non-genetic Risk Factors in Parkinson's Disease. Front Aging Neurosci 2020; 12:12. [PMID: 32226375 PMCID: PMC7080700 DOI: 10.3389/fnagi.2020.00012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
The exact cause of Parkinson’s disease (PD), the second most prevalent neurodegenerative disease in modern societies, is still unknown. Many scientists point out that PD is caused by a complex interaction between different factors. Although the main risk factor is age, there are other influences, genetic and environmental, that individually or in combination may trigger neurodegenerative changes leading to PD. Nowadays, research remains focused on better understanding which environmental factors are related to the risk of developing PD and why. In line with the knowledge on evidence on exposures that prevent/delay PD onset or that impact on disease progression, the aims of this review were: (i) to comment on the non-genetic risk factors that mainly affect idiopathic PD; and (ii) to comment on seemingly reliable preventive interventions. We discuss both environmental factors that may affect the central nervous system (CNS) or the intestinal tract, and the likely mechanisms underlying noxious or protective actions. Knowledge on risk, protective factors, and mechanisms may help to envisage why nigral dopaminergic neurons are so vulnerable in PD and, eventually, to design new strategies for PD prevention and/or anti-PD therapy. This article reviews the variety of the known and suspected environmental factors, such as lifestyle, gut microbiota or pesticide exposition, and distinguishes between those that are harmful or beneficial for the PD acquisition or progression. In fact, the review covers one of the most novel players in the whole picture, and we address the role of microbiota on keeping a healthy CNS and/or on preventing the “side-effects” related to aging.
Collapse
Affiliation(s)
- Rafael Franco
- Chemistry School, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Rivas-Santisteban
- Chemistry School, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, Madrid, Spain.,Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - Eva Martínez-Pinilla
- Departamento de Morfología y Biología Celular, Facultad de Medicina, Universidad de Oviedo, Oviedo, Spain.,Instituto de Neurociencias del Principado de Asturias (INEUROPA), Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
33
|
Neuroprotective effect of crocin against rotenone-induced Parkinson's disease in rats: Interplay between PI3K/Akt/mTOR signaling pathway and enhanced expression of miRNA-7 and miRNA-221. Neuropharmacology 2020; 164:107900. [DOI: 10.1016/j.neuropharm.2019.107900] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 10/14/2019] [Accepted: 11/30/2019] [Indexed: 11/23/2022]
|
34
|
Zilocchi M, Fasano M, Alberio T. Mitochondrial Proteins in the Development of Parkinson’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1158:17-44. [DOI: 10.1007/978-981-13-8367-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
35
|
Hinkle JT, Dawson VL, Dawson TM. The A1 astrocyte paradigm: New avenues for pharmacological intervention in neurodegeneration. Mov Disord 2019; 34:959-969. [PMID: 31136698 PMCID: PMC6642014 DOI: 10.1002/mds.27718] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/27/2019] [Accepted: 04/24/2019] [Indexed: 12/25/2022] Open
Abstract
We recently demonstrated that NLY01, a novel glucagon-like peptide-1 receptor agonist, exerts neuroprotective effects in two mouse models of PD in a glia-dependent manner. NLY01 prevented microglia from releasing inflammatory mediators known to convert astrocytes into a neurotoxic A1 reactive subtype. Importantly, we provided evidence that this neuroprotection was not mediated by a direct action of NLY01 on neurons or astrocytes (e.g., by activating neurotrophic pathways or modulating astrocyte reactivity per se). In the present article, we provide a generalist review of microglia and astrocytes in neurodegeneration and discuss the emerging paradigm of A1 astrocyte neurotoxicity in more detail. We comment on specific inferences that are naturally suggested by our work in this area and the differential level of support it offers to each. Finally, we discuss implications for the overall goal of creating disease-modifying therapies for PD, survey emerging methodologies for accelerating translational research on glia in neurodegeneration, and describe expected challenges for developing glia-directed therapies that do not impede essential physiological functions carried out by glia in the CNS. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jared T. Hinkle
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valina L. Dawson
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology,Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130-2685, USA
| | - Ted M. Dawson
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology,Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
36
|
Obata T. Glutaminergic tonic action potentiate MPP+-induced hydroxyl radical production in rat striatum. Neurosci Lett 2019; 705:51-53. [DOI: 10.1016/j.neulet.2019.02.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/03/2019] [Accepted: 02/09/2019] [Indexed: 11/15/2022]
|
37
|
Kurosaki H, Yamaguchi K, Man-Yoshi K, Muramatsu SI, Hara S, Ichinose H. Administration of tetrahydrobiopterin restored the decline of dopamine in the striatum induced by an acute action of MPTP. Neurochem Int 2019; 125:16-24. [PMID: 30739038 DOI: 10.1016/j.neuint.2019.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/19/2019] [Accepted: 02/06/2019] [Indexed: 10/27/2022]
Abstract
Parkinson's disease (PD) is the second common neurodegenerative disorder. Deficit of the nigro-striatal dopaminergic neurons causes the motor symptoms of PD. While the oxidative stress is thought to be deeply involved in the etiology of PD, molecular targets for the oxidative insults has not been fully elucidated. 6R-5,6,7,8-Tetrahydrobiopterin (BH4) is a cofactor for tyrosine hydroxylase (TH), the rate-limiting enzyme for production of dopamine, and easily oxidized to its dihydro-form. In this study, we examined the alteration in the metabolism of BH4 caused by a parkinsonian neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). MPTP reduced the dopamine content and the in vivo activity of TH in the striatum prior to degeneration of the dopaminergic neurons. We found that administration of BH4 could restore the dopamine content and in vivo TH activity in the striatum of MPTP-treated mice. Unexpectedly, when BH4 was administered with MPTP, BH4 contents in the brain were far higher than those injected without MPTP even at 23 h after the last injection. Because MPTP has been shown to increase ROS production in the dopaminergic neurons, we assumed that the increased ROS oxidizes BH4 into its dihydro-form, excreted from the dopaminergic neurons, taken-up by the neighboring cells, reduced back to BH4, and then accumulated in the brain. We also investigated the action of MPTP in mice lacking quinonoid-dihydropteridine reductase (Qdpr), an enzyme catalyzing regeneration of BH4 from quinonoid dihydrobiopterin. The dopamine depletion induced by MPTP was severer in Qdpr-deficient mice than in wild-type mice. The present data suggest that perturbation of the BH4 metabolism would be the cause of early and persistent dopamine depletion in the striatum.
Collapse
Affiliation(s)
- Hiroki Kurosaki
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Kentaro Yamaguchi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Kohei Man-Yoshi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Shin-Ichi Muramatsu
- Division of Neurology, Jichi Medical University, Shimotsuke, Tochigi, Japan; Center for Gene & Cell Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Satoshi Hara
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Hiroshi Ichinose
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.
| |
Collapse
|
38
|
Sbodio JI, Snyder SH, Paul BD. Redox Mechanisms in Neurodegeneration: From Disease Outcomes to Therapeutic Opportunities. Antioxid Redox Signal 2019; 30:1450-1499. [PMID: 29634350 PMCID: PMC6393771 DOI: 10.1089/ars.2017.7321] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 03/16/2018] [Accepted: 03/18/2018] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Once considered to be mere by-products of metabolism, reactive oxygen, nitrogen and sulfur species are now recognized to play important roles in diverse cellular processes such as response to pathogens and regulation of cellular differentiation. It is becoming increasingly evident that redox imbalance can impact several signaling pathways. For instance, disturbances of redox regulation in the brain mediate neurodegeneration and alter normal cytoprotective responses to stress. Very often small disturbances in redox signaling processes, which are reversible, precede damage in neurodegeneration. Recent Advances: The identification of redox-regulated processes, such as regulation of biochemical pathways involved in the maintenance of redox homeostasis in the brain has provided deeper insights into mechanisms of neuroprotection and neurodegeneration. Recent studies have also identified several post-translational modifications involving reactive cysteine residues, such as nitrosylation and sulfhydration, which fine-tune redox regulation. Thus, the study of mechanisms via which cell death occurs in several neurodegenerative disorders, reveal several similarities and dissimilarities. Here, we review redox regulated events that are disrupted in neurodegenerative disorders and whose modulation affords therapeutic opportunities. CRITICAL ISSUES Although accumulating evidence suggests that redox imbalance plays a significant role in progression of several neurodegenerative diseases, precise understanding of redox regulated events is lacking. Probes and methodologies that can precisely detect and quantify in vivo levels of reactive oxygen, nitrogen and sulfur species are not available. FUTURE DIRECTIONS Due to the importance of redox control in physiologic processes, organisms have evolved multiple pathways to counteract redox imbalance and maintain homeostasis. Cells and tissues address stress by harnessing an array of both endogenous and exogenous redox active substances. Targeting these pathways can help mitigate symptoms associated with neurodegeneration and may provide avenues for novel therapeutics. Antioxid. Redox Signal. 30, 1450-1499.
Collapse
Affiliation(s)
- Juan I. Sbodio
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Solomon H. Snyder
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bindu D. Paul
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
39
|
Togashi K, Hasegawa M, Nagai J, Tonouchi A, Masukawa D, Hensley K, Goshima Y, Ohshima T. Genetic suppression of collapsin response mediator protein 2 phosphorylation improves outcome in methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine‐induced Parkinson’s model mice. Genes Cells 2018; 24:31-40. [DOI: 10.1111/gtc.12651] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/18/2018] [Accepted: 10/18/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Kentaro Togashi
- Department of Life Science and Medical Bio‐Science Waseda University Tokyo Japan
| | - Masaya Hasegawa
- Department of Life Science and Medical Bio‐Science Waseda University Tokyo Japan
| | - Jun Nagai
- Department of Life Science and Medical Bio‐Science Waseda University Tokyo Japan
- Japan Society for the Promotion of Science Tokyo Japan
| | - Aine Tonouchi
- Department of Life Science and Medical Bio‐Science Waseda University Tokyo Japan
| | - Daiki Masukawa
- Department of Molecular Pharmacology and Neurobiology Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Kenneth Hensley
- Department of Biochemistry, Molecular and Cell Science Arkansas College of Osteopathic Medicine (ARCOM) Fort Smith Arkansas
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bio‐Science Waseda University Tokyo Japan
| |
Collapse
|
40
|
Choi YG, Hong YM, Kim LH, Yeo S, Lim S. Moutan Cortex Radicis inhibits the nigrostriatal damage in a 6-OHDA-induced Parkinson's disease model. Chin J Nat Med 2018; 16:490-498. [PMID: 30080647 DOI: 10.1016/s1875-5364(18)30084-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Indexed: 10/28/2022]
Abstract
The traditionally used oriental herbal medicine Moutan Cortex Radicis [MCR; Paeonia Suffruticosa Andrews (Paeoniaceae)] exerts anti-inflammatory, anti-spasmodic, and analgesic effects. In the present study, we investigated the therapeutic effects of differently fractioned MCR extracts in a 6-hydroxydopamine (OHDA)-induced Parkinson's disease model and neuro-blastoma B65 cells. Ethanol-extracted MCR was fractionated by n-hexane, butanol, and distilled water. Adult Sprague-Dawley rats were treated first with 20 μg of 6-OHDA, followed by three MCR extract fractions (100 or 200 mg·kg-1) for 14 consecutive days. In the behavioral rotation experiment, the MCR extract-treated groups showed significantly decreased number of net turns compared with the 6-OHDA control group. The three fractions also significantly inhibited the reduction in tyrosine hydroxylase-positive cells in the substantia nigra pars compacta following 6-OHDA neurotoxicity. Western blotting analysis revealed significantly reduced tyrosine hydroxylase expression in the substantia nigra pars compacta in the 6-OHDA-treated group, which was significantly inhibited by the n-hexane or distilled water fractions of MCR. B65 cells were exposed to the extract fractions for 24 h prior to addition of 6-OHDA for 30 min; treatment with n-hexane or distilled water fractions of MCR reduced apoptotic cell death induced by 6-OHDA neurotoxicity and inhibited nitric oxide production and neuronal nitric oxide synthase expression. These results showed that n-hexane- and distilled water-fractioned MCR extracts inhibited 6-OHDA-induced neurotoxicity by suppressing nitric oxide production and neuronal nitric oxide synthase activity, suggesting that MCR extracts could serve as a novel candidate treatment for the patients with Parkinson's disease.
Collapse
Affiliation(s)
- Yeong-Gon Choi
- Laboratory of Neurodegenerative Diseases, Ilsong Institute of Life Science, Hallym University, Anyang 14066, Republic of Korea; Research Group of Pain and Neuroscience, WHO Collaborating Center for Traditional Medicine, East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yeon-Mi Hong
- Research Group of Pain and Neuroscience, WHO Collaborating Center for Traditional Medicine, East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Applied Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Li-Hua Kim
- Research Group of Pain and Neuroscience, WHO Collaborating Center for Traditional Medicine, East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Applied Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sujung Yeo
- Research Group of Pain and Neuroscience, WHO Collaborating Center for Traditional Medicine, East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Meridian and Acupoint, College of Korean Medicine, Sang Ji University, Wonju 26339, Republic of Korea
| | - Sabina Lim
- Research Group of Pain and Neuroscience, WHO Collaborating Center for Traditional Medicine, East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Applied Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
41
|
Role of neuronal nitric oxide synthase in slowly progressive dopaminergic neurodegeneration in the Zitter rat. Nitric Oxide 2018; 78:41-50. [DOI: 10.1016/j.niox.2018.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/02/2018] [Accepted: 05/20/2018] [Indexed: 12/21/2022]
|
42
|
Zhang J, Culp ML, Craver JG, Darley-Usmar V. Mitochondrial function and autophagy: integrating proteotoxic, redox, and metabolic stress in Parkinson's disease. J Neurochem 2018; 144:691-709. [PMID: 29341130 PMCID: PMC5897151 DOI: 10.1111/jnc.14308] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/04/2018] [Accepted: 01/09/2018] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a movement disorder with widespread neurodegeneration in the brain. Significant oxidative, reductive, metabolic, and proteotoxic alterations have been observed in PD postmortem brains. The alterations of mitochondrial function resulting in decreased bioenergetic health is important and needs to be further examined to help develop biomarkers for PD severity and prognosis. It is now becoming clear that multiple hits on metabolic and signaling pathways are likely to exacerbate PD pathogenesis. Indeed, data obtained from genetic and genome association studies have implicated interactive contributions of genes controlling protein quality control and metabolism. For example, loss of key proteins that are responsible for clearance of dysfunctional mitochondria through a process called mitophagy has been found to cause PD, and a significant proportion of genes associated with PD encode proteins involved in the autophagy-lysosomal pathway. In this review, we highlight the evidence for the targeting of mitochondria by proteotoxic, redox and metabolic stress, and the role autophagic surveillance in maintenance of mitochondrial quality. Furthermore, we summarize the role of α-synuclein, leucine-rich repeat kinase 2, and tau in modulating mitochondrial function and autophagy. Among the stressors that can overwhelm the mitochondrial quality control mechanisms, we will discuss 4-hydroxynonenal and nitric oxide. The impact of autophagy is context depend and as such can have both beneficial and detrimental effects. Furthermore, we highlight the potential of targeting mitochondria and autophagic function as an integrated therapeutic strategy and the emerging contribution of the microbiome to PD susceptibility.
Collapse
Affiliation(s)
- Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
- Department of Veterans Affairs, Birmingham VA Medical Center
| | - M Lillian Culp
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| | - Jason G Craver
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| | - Victor Darley-Usmar
- Center for Free Radical Biology, University of Alabama at Birmingham
- Department of Pathology, University of Alabama at Birmingham
| |
Collapse
|
43
|
Potential neuroprotective effect of androst‐5‐ene‐3β, 17β‐diol (ADIOL) on the striatum, and substantia nigra in Parkinson's disease rat model. J Cell Physiol 2018; 233:5981-6000. [DOI: 10.1002/jcp.26412] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 12/19/2017] [Indexed: 12/14/2022]
|
44
|
Terron A, Bal-Price A, Paini A, Monnet-Tschudi F, Bennekou SH, Leist M, Schildknecht S. An adverse outcome pathway for parkinsonian motor deficits associated with mitochondrial complex I inhibition. Arch Toxicol 2018; 92:41-82. [PMID: 29209747 PMCID: PMC5773657 DOI: 10.1007/s00204-017-2133-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022]
Abstract
Epidemiological studies have observed an association between pesticide exposure and the development of Parkinson's disease, but have not established causality. The concept of an adverse outcome pathway (AOP) has been developed as a framework for the organization of available information linking the modulation of a molecular target [molecular initiating event (MIE)], via a sequence of essential biological key events (KEs), with an adverse outcome (AO). Here, we present an AOP covering the toxicological pathways that link the binding of an inhibitor to mitochondrial complex I (i.e., the MIE) with the onset of parkinsonian motor deficits (i.e., the AO). This AOP was developed according to the Organisation for Economic Co-operation and Development guidelines and uploaded to the AOP database. The KEs linking complex I inhibition to parkinsonian motor deficits are mitochondrial dysfunction, impaired proteostasis, neuroinflammation, and the degeneration of dopaminergic neurons of the substantia nigra. These KEs, by convention, were linearly organized. However, there was also evidence of additional feed-forward connections and shortcuts between the KEs, possibly depending on the intensity of the insult and the model system applied. The present AOP demonstrates mechanistic plausibility for epidemiological observations on a relationship between pesticide exposure and an elevated risk for Parkinson's disease development.
Collapse
Affiliation(s)
| | | | - Alicia Paini
- European Commission Joint Research Centre, Ispra, Italy
| | | | | | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstr. 10, PO Box M657, 78457, Konstanz, Germany
| | - Stefan Schildknecht
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstr. 10, PO Box M657, 78457, Konstanz, Germany.
| |
Collapse
|
45
|
Luo Y, Liu J, Sun X, Feng T, Fang L, Chen S, Fang C, Feng X, Huang H. Tsc1-dependent transcriptional programming of dendritic cell homeostasis and function. Exp Cell Res 2017; 363:73-83. [PMID: 29294307 DOI: 10.1016/j.yexcr.2017.12.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/21/2017] [Accepted: 12/29/2017] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DCs) are pivotal to initiating adaptive immune response. Emerging evidence highlights important roles of tuberous sclerosis complex 1 (Tsc1) in DC development and activation. Our previous study also showed that Tsc1 expression in DCs was required to promote T-cell homeostasis and response partially through inhibiting mammalian target of rapamycin complex1 (mTORC1). However, the molecular mechanism of transcriptional regulation by which Tsc1 control DC homeostasis and function remains largely unknown. Here we globally identified the Tsc1-regulated genes by comparing the transcriptional profiling of Tsc1-deficient DCs with wild-type DCs. It showed that Tsc1 specifically regulated the expression of groups of gene sets critically involved in DC survival, proliferation, metabolism and antigen presentation. The impacts of Tsc1 on DC gene expression were partially dependent on inhibition of mTORC1 signal. Our study thus provides a comprehensive molecular basis for understanding how Tsc1 programs the homeostasis and function of DCs through transcriptional regulation.
Collapse
Affiliation(s)
- Yuechen Luo
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Jingru Liu
- Central Laboratory, The Union Hospital of Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China
| | - Xiaolei Sun
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Tiantian Feng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Lijun Fang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Song Chen
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Chunmin Fang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China.
| | - Huifang Huang
- Central Laboratory, The Union Hospital of Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China.
| |
Collapse
|
46
|
Díaz-Casado ME, Rusanova I, Aranda P, Fernández-Ortiz M, Sayed RKA, Fernández-Gil BI, Hidalgo-Gutiérrez A, Escames G, López LC, Acuña-Castroviejo D. In Vivo Determination of Mitochondrial Respiration in 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Treated Zebrafish Reveals the Efficacy of Melatonin in Restoring Mitochondrial Normalcy. Zebrafish 2017; 15:15-26. [PMID: 29185873 DOI: 10.1089/zeb.2017.1479] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Although mitochondria dysfunction is related to multiple diseases, no in vivo studies are available on mitochondrial respiration in animal parkinsonian models. Our aim is to analyze in vivo mitochondrial respiration, which reflects changes in mitochondrial bioenergetics more precisely than in vitro mitochondrial preparations. These experiments can be carried out in zebrafish embryos, which were treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) from 24 to 72 hours postfertilization (hpf). A reduction in electron transfer system capacity, ATP turnover, and increased proton leak were observed at 72 hpf in MPTP-treated embryos. These changes were followed by a significant oxidative stress due to inhibition in antioxidative defense and autophagy impairment. After removing MPTP from the treatment at 72 hpf, these bioenergetic deficiencies persisted up to 120 hpf. The administration of melatonin to zebrafish embryos at 72 hpf, when mitochondrial dysfunction is already present, restored the respiratory capacity and ATP production, reduced oxidative stress, and normalized autophagy after 48 h. Melatonin also counteracted mortality and embryonic malformations due to MPTP. Our results confirm for the first time the efficacy of melatonin in restoring parkinsonian phenotypes in animals.
Collapse
Affiliation(s)
- María E Díaz-Casado
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,2 Departamento de Fisiología, Facultad de Medicina, Universidad de Granada , Granada, Spain
| | - Iryna Rusanova
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,2 Departamento de Fisiología, Facultad de Medicina, Universidad de Granada , Granada, Spain
| | - Paula Aranda
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,2 Departamento de Fisiología, Facultad de Medicina, Universidad de Granada , Granada, Spain
| | - Marisol Fernández-Ortiz
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,2 Departamento de Fisiología, Facultad de Medicina, Universidad de Granada , Granada, Spain
| | - Ramy K A Sayed
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,3 Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University , Sohag, Egypt
| | - Beatriz I Fernández-Gil
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,2 Departamento de Fisiología, Facultad de Medicina, Universidad de Granada , Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,2 Departamento de Fisiología, Facultad de Medicina, Universidad de Granada , Granada, Spain
| | - Germaine Escames
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,2 Departamento de Fisiología, Facultad de Medicina, Universidad de Granada , Granada, Spain
| | - Luis C López
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,2 Departamento de Fisiología, Facultad de Medicina, Universidad de Granada , Granada, Spain
| | - Darío Acuña-Castroviejo
- 1 Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada , Granada, Spain .,2 Departamento de Fisiología, Facultad de Medicina, Universidad de Granada , Granada, Spain .,4 Unidad de Gestión Clínica de Laboratorios Clínicos, CIBER de Fragilidad y Envejecimiento, Ibs.Granada, Complejo Hospitalario de Granada , Granada, Spain
| |
Collapse
|
47
|
Lieberman OJ, Choi SJ, Kanter E, Saverchenko A, Frier MD, Fiore GM, Wu M, Kondapalli J, Zampese E, Surmeier DJ, Sulzer D, Mosharov EV. α-Synuclein-Dependent Calcium Entry Underlies Differential Sensitivity of Cultured SN and VTA Dopaminergic Neurons to a Parkinsonian Neurotoxin. eNeuro 2017; 4:ENEURO.0167-17.2017. [PMID: 29177188 PMCID: PMC5701296 DOI: 10.1523/eneuro.0167-17.2017] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 11/01/2017] [Accepted: 11/09/2017] [Indexed: 12/27/2022] Open
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disease characterized by a loss of dopaminergic neurons in the substantia nigra (SN). Although mitochondrial dysfunction and dysregulated α-synuclein (aSyn) expression are postulated to play a role in PD pathogenesis, it is still debated why neurons of the SN are targeted while neighboring dopaminergic neurons of the ventral tegmental area (VTA) are spared. Using electrochemical and imaging approaches, we investigated metabolic changes in cultured primary mouse midbrain dopaminergic neurons exposed to a parkinsonian neurotoxin, 1-methyl-4-phenylpyridinium (MPP+). We demonstrate that the higher level of neurotoxicity in SN than VTA neurons was due to SN neuron-specific toxin-induced increase in cytosolic dopamine (DA) and Ca2+, followed by an elevation of mitochondrial Ca2+, activation of nitric oxide synthase (NOS), and mitochondrial oxidation. The increase in cytosolic Ca2+ was not caused by MPP+-induced oxidative stress, but rather depended on the activity of both L-type calcium channels and aSyn expression, suggesting that these two established pathogenic factors in PD act in concert.
Collapse
Affiliation(s)
- Ori J. Lieberman
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Se Joon Choi
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Ellen Kanter
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Anastasia Saverchenko
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Micah D. Frier
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Giulia M. Fiore
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Min Wu
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| | - Jyothisri Kondapalli
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Enrico Zampese
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - D. James Surmeier
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - David Sulzer
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
- Department of Pharmacology, Columbia University Medical Center, New York, NY 10032
| | - Eugene V. Mosharov
- New York State Psychiatric Institute, Columbia University Medical Center, New York, NY 10032
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
48
|
Zhu Z, Yi S, Shan Z, Guo H, Ke S. Effect of isoflurane + N 2O inhalation and propofol + fentanyl anesthesia on myocardial function as assessed by cardiac troponin, caspase-3, cyclooxygenase-2 and inducible nitric oxide synthase expression. Exp Ther Med 2017; 14:4377-4382. [PMID: 29067116 DOI: 10.3892/etm.2017.5029] [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: 02/21/2016] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to evaluate the effect of isoflurane + N2O inhalation and propofol + fentanyl anesthesia on myocardial function as assessed by cardiac troponin T (cTnT). A total of 60 patients were randomized into two groups: isoflurane + N2O inhalation (n=30) and propofol + fentanyl anesthesia (n=30). The findings demonstrated that there was no significant difference between the two experimental groups in terms of cTnT levels, demographic properties or hemodynamic parameters. Isoflurane + N2O inhalation and propofol + fentanyl anesthesia, respectively, were also investigated in a rat model of myocardial infarction. Myocardial cell damage, inflammation and oxidative stress levels, caspase-3/9 activities and cyclooxygenase-2 protein expression were markedly decreased, although there was no statistical significance difference between the two experimental groups. Notably, inducible nitric oxide synthase protein expression in the isoflurane + N2O inhalation group was significantly higher than that of the propofol + fentanyl anesthesia group (P<0.01). In conclusion, isoflurane + N2O inhalation and propofol + fentanyl anesthesia are not associated with risks for myocardial function.
Collapse
Affiliation(s)
- Zhuanghui Zhu
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Shuanglian Yi
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhonggui Shan
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China.,Department of Cardiac Surgery, Teaching Hospital of Fujian Medical University, Xiamen, Fujian 361003, P.R. China
| | - Hongwei Guo
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Shaofan Ke
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| |
Collapse
|
49
|
Dawson TM, Dawson VL. Nitric Oxide Signaling in Neurodegeneration and Cell Death. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:57-83. [PMID: 29413528 DOI: 10.1016/bs.apha.2017.09.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this tribute to Solomon H. Snyder (Sol) we discuss the mechanisms by which nitric oxide (NO) kills neurons. We provide a historical perspective regarding the discovery that glutamate excitotoxicity is mediated by NO. It also contains a discussion of the discovery that neuronal nitric oxide synthase (nNOS) catalytic activity accounts for NADPH diaphorase activity and its localization in the central nervous system. NADPH diaphorase/nNOS neurons are unique in that they are resistant to toxic effects of excess glutamate and that they are resistant to neurodegeneration in a variety of neurodegenerative diseases. NADPH diaphorase/nNOS neurons are resistant to neurotoxicity and neurodegeneration through the overexpression of manganese superoxide dismutase. The review also delves into the mechanisms by which NO kills neurons including NO's activation of the glyceraldehyde-3-phosphate dehydrogenase-dependent cell pathway. In addition, there is a review of parthanatos in which NO combines with the superoxide anion ( [Formula: see text] ) to form peroxynitrite (ONOO-) that damages DNA and activates poly (ADP-ribose) (PAR) polymerase (PARP). This ultimately leads to activation of the PARP-dependent apoptosis-inducing factor-associated nuclease, the final executioner in NO-dependent cell death. Finally, there is a discussion of potential targets that are under development that target the mechanisms by which NO kills neurons.
Collapse
Affiliation(s)
- Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Johns Hopkins University School of Medicine, Baltimore, MD, United States; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, United States; Diana Helis Henry Medical Research Foundation, New Orleans, LA, United States.
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Johns Hopkins University School of Medicine, Baltimore, MD, United States; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, United States; Diana Helis Henry Medical Research Foundation, New Orleans, LA, United States.
| |
Collapse
|
50
|
Patel M, McElroy PB. Mitochondrial Dysfunction in Parkinson’s Disease. OXIDATIVE STRESS AND REDOX SIGNALLING IN PARKINSON’S DISEASE 2017. [DOI: 10.1039/9781782622888-00061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disorders where oxidative stress and mitochondrial dysfunction have been implicated as etiological factors. Mitochondria are the major producers of reactive oxygen species (ROS) that can have damaging effects to cellular macromolecules leading to neurodegeneration. The most compelling evidence for the role of mitochondria in the pathogenesis of PD has been derived from toxicant-induced models of parkinsonism. Over the years, epidemiological studies have suggested a link between exposure to environmental toxins such as pesticides and the risk of developing PD. Data from human and experimental studies involving the use of chemical agents like paraquat, diquat, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, rotenone and maneb have provided valuable insight into the underlying mitochondrial mechanisms contributing to PD and associated neurodegeneration. In this review, we have discussed the role of mitochondrial ROS and dysfunction in the pathogenesis of PD with a special focus on environmental agent-induced parkinsonism. We have described the various mitochondrial mechanisms by which such chemicals exert neurotoxicity, highlighting some landmark epidemiological and experimental studies that support the role of mitochondrial ROS and oxidative stress in contributing to these effects. Finally, we have discussed the significance of these studies in understanding the mechanistic underpinnings of PD-related dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| | - Pallavi Bhuyan McElroy
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| |
Collapse
|