1
|
Belviranlı M, Okudan N, Sezer T. Potential therapeutic effects of curcumin, with or without L-DOPA, on motor and cognitive functions and hippocampal changes in rotenone-treated rats. Metab Brain Dis 2025; 40:174. [PMID: 40208367 PMCID: PMC11985604 DOI: 10.1007/s11011-025-01602-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
The neurodegenerative condition known as Parkinson's disease (PD) is a long-term condition that causes both motor and non-motor symptoms. It is known that curcumin has a strong neuroprotective potential. This experimental study was designed to examine the anti-inflammatory, anti-apoptotic and neuroprotective effects of curcumin administered alone and in combination with L-DOPA in the hippocampus as well as behavioral symptoms in rotenone-induced PD model. Forty-two 4-month-old adult male Wistar rats were randomly divided into six groups as follows: Control, Curcumin, Rotenone, Rotenone plus curcumin, Rotenone plus L-DOPA and Rotenone plus curcumin plus L-DOPA. Control group received vehicles, curcumin group received curcumin (200 mg kg-1, daily for 35 days), rotenone group received rotenone (2 mg kg-1, daily for 35 days), and test groups received curcumin or L-DOPA (10 mg kg-1, daily for the last 15 days) or their combination in addition the rotenone. Pole, sucrose preference, open field, elevated plus maze, and Morris water maze tests were performed after treatment. Molecular and biochemical analyses were performed in the hippocampus tissue and serum samples. Rotenone injection caused impairments in motor activity, depressive-like behavior, and learning and memory functions. Rotenone also increased the expressions of α-synuclein, caspase 3, NF-κB, and decreased the expressions of parkin and BDNF in the hippocampus. However, especially curcumin and L-DOPA combined treatment normalized all these impaired molecular and behavioral variables. In conclusion, curcumin may exert beneficial effects in treatment strategies for PD-related hippocampal effects, especially when added to L-DOPA therapy.
Collapse
Affiliation(s)
- Muaz Belviranlı
- School of Medicine, Department of Physiology, Selçuk University, Konya, 42131, Turkey.
| | - Nilsel Okudan
- School of Medicine, Department of Physiology, Selçuk University, Konya, 42131, Turkey
| | - Tuğba Sezer
- School of Medicine, Department of Physiology, Selçuk University, Konya, 42131, Turkey
| |
Collapse
|
2
|
Ebadpour N, Mahmoudi M, Kamal Kheder R, Abavisani M, Baridjavadi Z, Abdollahi N, Esmaeili SA. From mitochondrial dysfunction to neuroinflammation in Parkinson's disease: Pathogenesis and mitochondrial therapeutic approaches. Int Immunopharmacol 2024; 142:113015. [PMID: 39222583 DOI: 10.1016/j.intimp.2024.113015] [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: 02/28/2024] [Revised: 07/28/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Parkinson's disease (PD) is a prevalent and intricate neurological condition resulting from a combination of several factors, such as genetics, environment, and the natural process of aging. Degeneration of neurons in the substantia nigra pars compacta (SN) can cause motor and non-motor impairments in patients with PD. In PD's etiology, inflammation and mitochondrial dysfunction play significant roles in the disease's development. Studies of individuals with PD have revealed increased inflammation in various brain areas. Furthermore, mitochondrial dysfunction is an essential part of PD pathophysiology. Defects in the components of the mitochondrial nucleus, its membrane or internal signaling pathways, mitochondrial homeostasis, and morphological alterations in peripheral cells have been extensively documented in PD patients. According to these studies, neuroinflammation and mitochondrial dysfunction are closely connected as pathogenic conditions in neurodegenerative diseases like PD. Given the mitochondria's role in cellular homeostasis maintenance in response to membrane structural flaws or mutations in mitochondrial DNA, their dynamic nature may present therapeutic prospects in this area. Recent research investigates mitochondrial transplantation as a potential treatment for Parkinson's disease in damaged neurons. This review delves into the impact of inflammation and mitochondrial dysfunction on PD occurrence, treatment approaches, and the latest developments in mitochondrial transplantation, highlighting the potential consequences of these discoveries.
Collapse
Affiliation(s)
- Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramiar Kamal Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania, Sulaymaniyah, Iraq; Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Baridjavadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Abdollahi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Xu C, Tang Y, Lu X, Chen R. Fyn, an important molecule in the brain, is a potential therapeutic target for brain tumours. Front Pharmacol 2024; 15:1485919. [PMID: 39697541 PMCID: PMC11652172 DOI: 10.3389/fphar.2024.1485919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
Under normal physiological conditions, Fyn, a nonreceptor tyrosine kinase, is involved in signal transduction pathways in the nervous system and in the formation and activation of T lymphocytes. Fyn is a member of the Src family of kinases (SFKs) and plays a role in cell morphogenic transformation, motility, proliferation, and death, which in turn influences the development and progression of various cancer types. SFKs are overexpressed or hyperactive in tumours, and they are engaged in several signalling pathways that lead to tumour development. Inhibition of Fyn can enhance patient outcomes and prolong survival. Thus, Fyn is a desirable therapeutic target in a variety of tumour types. To lay the groundwork for further investigation and targeted therapy in tumours, in this article, we review the most recent findings on the function of Fyn in tumours, with an emphasis on its role in gliomas. Understanding the function of Fyn during tumourigenesis and development and in resistance to anticancer therapeutic agents can aid in the development and application of innovative medicines that specifically target this kinase, thus improving the management of cancers.
Collapse
Affiliation(s)
- Chongxi Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Tang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xing Lu
- Department of Gynecological Nursing, West China Second Hospital, Sichuan University, Chengdu, China
| | - Ruiqi Chen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
K. A, Singh S. Neuro-inflammatory Responses in Alzheimer’s v/s Parkinson’s Diseases. ADVANCES IN DIAGNOSTICS AND IMMUNOTHERAPEUTICS FOR NEURODEGENERATIVE DISEASES 2024:17-31. [DOI: 10.2174/9789815238754124010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Neurodegenerative diseases are a heterogeneous group of disorders and are
the leading cause of morbidity and disability. These are described by the progressive
degeneration of the neurons and impaired function of the central nervous system.
Prevailing neurodegenerative diseases in the world include Alzheimer's disease and
Parkinson's disease and reports predict that on average, the prevalence of both diseases
will double in a span of the next twenty years. Pieces of evidence showed that the
immune system is profoundly involved in brain development, maintenance, and repair
as well as in damage, therefore, may provide a wide scope to focus on the
neuroinflammation-based therapeutic approaches. In this chapter, the various
neuroinflammatory responses will be discussed during the onset and progression of
both Alzheimer’s and Parkinson’s disease pathologies. We will be focusing on both
central and peripheral inflammatory responses and their consideration for disease
diagnosis and therapeutics.
Collapse
Affiliation(s)
- Amrutha K.
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute,
Lucknow-226031, India
| | - Sarika Singh
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
5
|
Zhang YY, Jiang XH, Zhu PP, Zhuo WY, Liu LB. Advancements in understanding substantia nigra hyperechogenicity via transcranial sonography in Parkinson's disease and its clinical implications. Front Neurol 2024; 15:1407860. [PMID: 39091976 PMCID: PMC11291319 DOI: 10.3389/fneur.2024.1407860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Amidst rising Parkinson's disease (PD) incidence in an aging global population, the need for non-invasive and reliable diagnostic methods is increasingly critical. This review evaluates the strategic role of transcranial sonography (TCS) in the early detection and monitoring of PD. TCS's ability to detect substantia nigra hyperechogenicity offers profound insights into its correlation with essential neuropathological alterations-namely, iron accumulation, neuromelanin depletion, and glial proliferation-fundamental to PD's pathophysiology. Our analysis highlights TCS's advantages, including its non-invasiveness, cost-effectiveness, and ease of use, positioning it as an invaluable tool for early diagnosis and continual disease progression monitoring. Moreover, TCS assists in identifying potential risk and protective factors, facilitating tailored therapeutic strategies to enhance clinical outcomes. This review advocates expanding TCS utilization and further research to maximize its diagnostic and prognostic potential in PD management, contributing to a more nuanced understanding of the disease.
Collapse
Affiliation(s)
- Yuan-yuan Zhang
- Department of Neurology, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| | - Xu-hong Jiang
- Department of Health Management, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| | - Pei-pei Zhu
- Department of Neurology, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| | - Wen-yan Zhuo
- Department of Neurology, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| | - Li-bin Liu
- Department of Neurology, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| |
Collapse
|
6
|
Bucher ML, Dicent J, Duarte Hospital C, Miller GW. Neurotoxicology of dopamine: Victim or assailant? Neurotoxicology 2024; 103:175-188. [PMID: 38857676 PMCID: PMC11694735 DOI: 10.1016/j.neuro.2024.06.001] [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: 04/13/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Since the identification of dopamine as a neurotransmitter in the mid-20th century, investigators have examined the regulation of dopamine homeostasis at a basic biological level and in human disorders. Genetic animal models that manipulate the expression of proteins involved in dopamine homeostasis have provided key insight into the consequences of dysregulated dopamine. As a result, we have come to understand the potential of dopamine to act as an endogenous neurotoxin through the generation of reactive oxygen species and reactive metabolites that can damage cellular macromolecules. Endogenous factors, such as genetic variation and subcellular processes, and exogenous factors, such as environmental exposures, have been identified as contributors to the dysregulation of dopamine homeostasis. Given the variety of dysregulating factors that impact dopamine homeostasis and the potential for dopamine itself to contribute to further cellular dysfunction, dopamine can be viewed as both the victim and an assailant of neurotoxicity. Parkinson's disease has emerged as the exemplar case study of dopamine dysregulation due to the genetic and environmental factors known to contribute to disease risk, and due to the evidence of dysregulated dopamine as a pathologic and pathogenic feature of the disease. This review, inspired by the talk, "Dopamine in Durham: location, location, location" presented by Dr. Miller for the Jacob Hooisma Memorial Lecture at the International Neurotoxicology Association meeting in 2023, offers a primer on dopamine toxicity covering endogenous and exogenous factors that disrupt dopamine homeostasis and the actions of dopamine as an endogenous neurotoxin.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Jocelyn Dicent
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Carolina Duarte Hospital
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
7
|
Suleiman Khoury Z, Sohail F, Wang J, Mendoza M, Raake M, Tahoor Silat M, Reddy Bathinapatta M, Sadeghzadegan A, Meghana P, Paul J. Neuroinflammation: A Critical Factor in Neurodegenerative Disorders. Cureus 2024; 16:e62310. [PMID: 39006715 PMCID: PMC11246070 DOI: 10.7759/cureus.62310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
This review offers a comprehensive review of the signals and the paramount role neuroinflammation plays in neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's, and amyotrophic lateral sclerosis. The study explores the sophisticated interactions between microglial, astrocytic, and dendritic cells and how neuroinflammation affects long-term neuronal damage and dysfunction. There are specific pathways related to the mentioned inflammatory processes, including Janus kinases/signal transducer and activator of transcriptions, nuclear factor-κB, and mitogen-activated protein kinases pathways. Neuroinflammation is argued to be a double-edged sword, being not only a protective agent that prevents further neuron damage but also the causative factor in more cell injury development. This concept of contrasting inflammation with neuroprotection advocates for the use of therapeutic techniques that seek to modulate neuroinflammatory responses as part of the neurodegeneration treatment. The recent research findings are integrated with the established knowledge to help present a comprehensive image of neuroinflammation's impact on neurodegenerative diseases and its implications for future therapy.
Collapse
Affiliation(s)
| | - Fatima Sohail
- Department of Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, USA
| | - Jada Wang
- Department of Medicine, St. George's University, Brooklyn, USA
| | - Moises Mendoza
- Department of Health Sciences, Universidad Centroccidental Lisandro Alvarado, Barquisimeto, VEN
| | - Mohammed Raake
- Department of Medicine, Annamalai University, Chennai, IND
| | | | | | - Amirali Sadeghzadegan
- Department of General Practice, Marmara University School of Medicine, Istanbul, TUR
| | - Patel Meghana
- Department of Medicine, Ramaiah University of Applied Sciences, Bengaluru, IND
| | - Janisha Paul
- Department of Medicine, Punjab Institute of Medical Sciences, Jalandhar, IND
| |
Collapse
|
8
|
Valvaikar S, Vaidya B, Sharma S, Bishnoi M, Kondepudi KK, Sharma SS. Supplementation of probiotic Bifidobacterium breve Bif11 reverses neurobehavioural deficits, inflammatory changes and oxidative stress in Parkinson's disease model. Neurochem Int 2024; 174:105691. [PMID: 38311217 DOI: 10.1016/j.neuint.2024.105691] [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: 11/04/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Human gut microbiota are thought to affect different physiological processes in the body, including brain functions. Gut dysbiosis has been linked to the progression of Parkinson's disease (PD) and thus, restoring the healthy gut microbiota with supplementation of putative probiotic strains can confer some benefits in PD. In the current study, we explored the neuroprotective potential of Bifidobacterium breve Bif11 supplementation in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) treated female Sprague Dawley rats. This study investigated the behavioural, molecular and biochemical parameters in the MPTP rat model. A pharmacological intervention of Bif11 at doses of 1 × 1010 CFU and 2 × 1010 CFU for 21 days was found to attenuate the cognitive and motor changes in the MPTP rat model. Furthermore, it also increased the tyrosine hydroxylase levels, reduced pro-inflammatory markers and decreased oxidative and nitrosative stress in the mid brain of MPTP-lesioned rats. Bif11 supplementation even restored the levels of short-chain fatty acids and decreased intestinal epithelial permeability in MPTP-induced PD model rats. In summary, these findings demonstrate that B. breve Bif11 has the potential to ameliorate symptoms of PD. However, this therapy needs to be further investigated with in-depth mechanistic insights in the future for the treatment of PD.
Collapse
Affiliation(s)
- Sonali Valvaikar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India
| | - Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India
| | - Shikha Sharma
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India
| | - Mahendra Bishnoi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India.
| | - Shyam S Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India.
| |
Collapse
|
9
|
Dias-Carvalho A, Sá SI, Carvalho F, Fernandes E, Costa VM. Inflammation as common link to progressive neurological diseases. Arch Toxicol 2024; 98:95-119. [PMID: 37964100 PMCID: PMC10761431 DOI: 10.1007/s00204-023-03628-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023]
Abstract
Life expectancy has increased immensely over the past decades, bringing new challenges to the health systems as advanced age increases the predisposition for many diseases. One of those is the burden of neurologic disorders. While many hypotheses have been placed to explain aging mechanisms, it has been widely accepted that the increasing pro-inflammatory status with advanced age or "inflammaging" is a main determinant of biological aging. Furthermore, inflammaging is at the cornerstone of many age-related diseases and its involvement in neurologic disorders is an exciting hypothesis. Indeed, aging and neurologic disorders development in the elderly seem to share some basic pathways that fundamentally converge on inflammation. Peripheral inflammation significantly influences brain function and contributes to the development of neurological disorders, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Understanding the role of inflammation in the pathogenesis of progressive neurological diseases is of crucial importance for developing effective treatments and interventions that can slow down or prevent disease progression, therefore, decreasing its social and economic burden.
Collapse
Affiliation(s)
- Ana Dias-Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO- Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| | - Susana Isabel Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO- Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO- Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
10
|
Alrouji M, Al-Kuraishy HM, Al-Mahammadawy AKAA, Al-Gareeb AI, Saad HM, Batiha GES. The potential role of cholesterol in Parkinson's disease neuropathology: perpetrator or victim. Neurol Sci 2023; 44:3781-3794. [PMID: 37428278 DOI: 10.1007/s10072-023-06926-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/13/2023] [Indexed: 07/11/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by deposition of α-synuclein and aggregation of Lewy bodies. Cholesterol is involved with PD neuropathology in bidirectional ways that could be protective or harmful. Thus, the objective of the present review was to verify the potential role of cholesterol in PD neuropathology. Deregulation of ion channels and receptors induced by cholesterol alteration suggests a possible mechanism for the neuroprotective effects of cholesterol against PD development. However, high serum cholesterol level increases PD risk indirectly by 27-hydroxycholesterol which induces oxidative stress, inflammation, and apoptosis. Besides, hypercholesterolemia triggers the accumulation of cholesterol in macrophages and immune cells leading to the release of pro-inflammatory cytokines with progression of neuroinflammation subsequently. Additionally, cholesterol increases aggregation of α-synuclein and induces degeneration of dopaminergic neurons (DN) in the substantia nigra (SN). Hypercholesterolemia may lead to cellular Ca2+ overload causing synaptic and the development of neurodegeneration. In conclusion, cholesterol has bidirectional effects on PD neuropathology and might be protective or harmful.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, Al-Mustansiriyah University, M.B.Ch.B, FRCP; Box, Baghdad, 14132, Iraq
| | | | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, Al-Mustansiriyah University, M.B.Ch.B, FRCP; Box, Baghdad, 14132, Iraq
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matrouh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Al Beheira, 22511, Egypt.
| |
Collapse
|
11
|
Omotayo T, Otenaike TA, Adedara AO, Adeyemi OE, Jonhnson TO, Abolaji AO. Biological interactions and attenuation of MPTP-induced toxicity in Drosophila melanogaster by Trans-astaxanthin. Neurosci Res 2023; 196:52-58. [PMID: 37329901 DOI: 10.1016/j.neures.2023.06.005] [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: 11/30/2022] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023]
Abstract
Trans-astaxanthin (TA) is a carotenoid with amphipathic chemical structure found in yeast, and aquatic organisms. It is known to possess both antioxidative and anti-inflammatory properties. This study was carried out to investigate the ameliorative action of TA on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced toxicity in Drosophila melanogaster (Fruit fly). The flies were orally treated with TA (2.5 mg/10 g diet) and/or MPTP (500 µM) for 5 days. Thereafter, we evaluated selected biomarkers of locomotor deficits (acetylcholinesterase (AChE) and negative geotaxis), oxidative stress (hydrogen peroxide (H2O2), protein carbonyls (PC)), antioxidants (total thiols (T-SH), non-protein thiols, glutathione-S-transferase (GST) and catalase), and inflammation (nitric oxide (nitrite/nitrate) in the flies. Furthermore, we investigated molecular docking analysis of TA against Kelch-like ECH-associated protein 1 (Keap1)) of Homo sapiens and D. melanogaster. The results indicated that TA increased MPTP-induced decreased activities of AChE, GST, and catalase, as well as levels of non-protein thiols and T-SH compared with MPTP-treated flies (p < 0.05). Furthermore, TA attenuated inflammation, and improved locomotor deficit in the flies. The molecular docking data showed that TA had docking scores for binding both the Human and Drosophila Keap1, nearly closer to or higher than the standard inhibitor. The attenuating effects of TA against MPTP-induced toxicity could arise from its antioxidative and anti-inflammatory properties as well as its chemical structure.
Collapse
Affiliation(s)
- Tolulope Omotayo
- Drosophila Laboratory. Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Titilayomi A Otenaike
- Drosophila Laboratory. Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeola Oluwatosin Adedara
- Drosophila Laboratory. Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria; Drosophila Research and Training Centre, Basorun, Ibadan, Nigeria
| | - Oluwagbenga Eyitayo Adeyemi
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, University of Jos, Jos, Nigeria
| | - Titilayo O Jonhnson
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, University of Jos, Jos, Nigeria
| | - Amos Olalekan Abolaji
- Drosophila Laboratory. Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria; Drosophila Research and Training Centre, Basorun, Ibadan, Nigeria.
| |
Collapse
|
12
|
Mohammad, Khan UA, Warsi MH, Alkreathy HM, Karim S, Jain GK, Ali A. Intranasal cerium oxide nanoparticles improves locomotor activity and reduces oxidative stress and neuroinflammation in haloperidol-induced parkinsonism in rats. Front Pharmacol 2023; 14:1188470. [PMID: 37324485 PMCID: PMC10267740 DOI: 10.3389/fphar.2023.1188470] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/09/2023] [Indexed: 08/26/2023] Open
Abstract
Introduction: Cerium oxide nanoparticles (CONPs) have been investigated for their therapeutic potential in Parkinson's disease (PD) due to their potent and regenerative antioxidant activity. In the present study, CONPs were used to ameliorate the oxidative stress caused by free radicals in haloperidol-induced PD in rats following intranasal administration. Method: The antioxidant potential of the CONPs was evaluated in vitro using ferric reducing antioxidant power (FRAP) assay. The penetration and local toxicity of the CONPs was evaluated ex-vivo using goat nasal mucosa. The acute local toxicity of intranasal CONPs was also studied in rat. Gamma scintigraphy was used to assess the targeted brain delivery of CONPs. Acute toxicity studies were performed in rats to demonstrate safety of intranasal CONPs. Further, open field test, pole test, biochemical estimations and brain histopathology was performed to evaluate efficacy of intranasal CONPs in haloperidol-induced PD rat model. Results: The FRAP assay revealed highest antioxidant activity of prepared CONPs at a concentration of 25 μg/mL. Confocal microscopy showed deep and homogenous distribution of CONPs in the goat nasal mucus layers. No signs of irritation or injury were seen in goat nasal membrane when treated with optimized CONPs. Scintigraphy studies in rats showed targeted brain delivery of intranasal CONPs and acute toxicity study demonstrated safety. The results of open field and pole test showed highly significant (p < 0.001) improvement in locomotor activity of rats treated with intranasal CONPs compared to untreated rats. Further, brain histopathology of treatment group rats showed reduced neurodegeneration with presence of more live cells. The amount of thiobarbituric acid reactive substances (TBARS) was reduced significantly, whereas the levels of catalase (CAT), superoxide dismutase (SOD), and GSH were increased significantly, while amounts of interleukin-6 (IL-6) and tumour necrosis factor-alpha (TNF-α) showed significant reduction after intranasal administration of CONPs. Also, the intranasal CONPs, significantly high (p < 0.001) dopamine concentration (13.93 ± 0.85 ng/mg protein) as compared to haloperidol-induced control rats (5.76 ± 0.70 ng/mg protein). Conclusion: The overall results concluded that the intranasal CONPs could be safe and effective therapeutics for the management of PD.
Collapse
Affiliation(s)
- Mohammad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Urooj Ahmed Khan
- Department of Pharmaceutics, Dr. Ram Manohar Lohia College of Pharmacy, Ghaziabad, Uttar Pradesh, India
| | - Musarrat Husain Warsi
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Huda Mohammed Alkreathy
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shahid Karim
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Gaurav Kumar Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
- Center for Advanced Formulation Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Asgar Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| |
Collapse
|
13
|
Kim EY, Kim JE, Kim YE, Choi B, Sohn DH, Park SO, Chung YH, Kim Y, Robinson WH, Kim YG, Chang EJ. Dysfunction in parkin aggravates inflammatory bone erosion by reinforcing osteoclast activity. Cell Biosci 2023; 13:48. [PMID: 36882866 PMCID: PMC9993703 DOI: 10.1186/s13578-023-00973-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/25/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Parkin dysfunction associated with the progression of parkinsonism contributes to a progressive systemic skeletal disease characterized by low bone mineral density. However, the role of parkin in bone remodeling has not yet been elucidated in detail. RESULT We observed that decreased parkin in monocytes is linked to osteoclastic bone-resorbing activity. siRNA-mediated knockdown of parkin significantly enhanced the bone-resorbing activity of osteoclasts (OCs) on dentin without any changes in osteoblast differentiation. Moreover, Parkin-deficient mice exhibited an osteoporotic phenotype with a lower bone volume accompanied by increased OC-mediated bone-resorbing capacity displaying increased acetylation of α-tubulin compared to wild-type (WT) mice. Notably, compared to WT mice, the Parkin-deficient mice displayed increased susceptibility to inflammatory arthritis, reflected by a higher arthritis score and a marked bone loss after arthritis induction using K/BxN serum transfer, but not ovariectomy-induced bone loss. Intriguingly, parkin colocalized with microtubules and parkin-depleted-osteoclast precursor cells (Parkin-/- OCPs) displayed augmented ERK-dependent acetylation of α-tubulin due to failure of interaction with histone deacetylase 6 (HDAC6), which was promoted by IL-1β signaling. The ectopic expression of parkin in Parkin-/- OCPs limited the increase in dentin resorption induced by IL-1β, accompanied by the reduced acetylation of α-tubulin and diminished cathepsin K activity. CONCLUSION These results indicate that a deficiency in the function of parkin caused by a decrease in parkin expression in OCPs under the inflammatory condition may enhance inflammatory bone erosion by altering microtubule dynamics to maintain OC activity.
Collapse
Affiliation(s)
- Eun-Young Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Ji-Eun Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Young-Eun Kim
- Department of Rheumatology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Bongkun Choi
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, 50612, Korea
| | - Si-On Park
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.,Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Yeon-Ho Chung
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Yongsub Kim
- Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.,Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yong-Gil Kim
- Department of Rheumatology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea. .,Stem Cell Immunomodulation Research Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
| |
Collapse
|
14
|
Batiha GES, Al-kuraishy HM, Al-Gareeb AI, Elekhnawy E. SIRT1 pathway in Parkinson's disease: a faraway snapshot but so close. Inflammopharmacology 2023; 31:37-56. [PMID: 36580159 PMCID: PMC9957916 DOI: 10.1007/s10787-022-01125-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 12/19/2022] [Indexed: 12/30/2022]
Abstract
Silent information regulator (SIRT) has distinctive enzymatic activities and physiological functions to control cell-cycle progression, gene expression, and DNA stability by targeting histone and non-histone proteins. SIRT1 enhances synaptic formation and synaptic activity, and therefore, can reduce the progression of various degenerative brain diseases including Parkinson's disease (PD). SIRT1 activity is decreased by aging with a subsequent increased risk for the development of degenerative brain diseases. Inhibition of SIRT1 promotes inflammatory reactions since SIRT1 inhibits transcription of nuclear factor kappa B (NF-κB) which also inhibits SIRT1 activation via activation of microRNA and miR-34a which reduce NAD synthesis. SIRT1 is highly expressed in microglia as well as neurons, and has antioxidant and anti-inflammatory effects. Therefore, this review aimed to find the possible role of SIRT1 in PD neuropathology. SIRT1 has neuroprotective effects; therefore, downregulation of SIRT1 during aging promotes p53 expression and may increase the vulnerability of neuronal cell deaths. PD neuropathology is linked with the sequence of inflammatory changes and the release of pro-inflammatory cytokines due to the activation of inflammatory signaling pathways. In addition, oxidative stress, inflammatory disorders, mitochondrial dysfunction, and apoptosis contribute mutually to PD neuropathology. Thus, SIRT1 and SIRT1 activators play a crucial role in the mitigation of PD neuropathology through the amelioration of oxidative stress, inflammatory disorders, mitochondrial dysfunction, apoptosis, and inflammatory signaling pathways.
Collapse
Affiliation(s)
- Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 AlBeheira Egypt
| | - Hayder M. Al-kuraishy
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132 Iraq
| | - Ali I. Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, 14132 Iraq
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527 Egypt
| |
Collapse
|
15
|
Repositioning doxycycline for treating synucleinopathies: Evidence from a pre-clinical mouse model. Parkinsonism Relat Disord 2023; 106:105229. [PMID: 36462409 DOI: 10.1016/j.parkreldis.2022.105229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/02/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Parkinson's disease remains orphan of valuable therapies capable to interfere with the disease pathogenesis despite the large number of symptomatic approaches adopted in clinical practice to manage this disease. Treatments simultaneously affecting α-synuclein (α-syn) oligomerization and neuroinflammation may counteract Parkinson's disease and related disorders. Recent data demonstrate that Doxycycline, a tetracycline antibiotic, can inhibit α-syn aggregation as well as neuroinflammation. We herein investigate, for the first time, the potential therapeutic properties of Doxy in a human α-syn A53T transgenic Parkinson's disease mouse model evaluating behavioural, biochemical and histopathological parameters. EXPERIMENTAL APPROACH Human α-syn A53T transgenic mice were treated with Doxycycline (10 mg/kg daily ip) for 30 days. The effect of treatment on motor, cognitive and daily live activity performances were examined. Neuropathological and neurophysiological parameters were assessed through immunocytochemical, electrophysiological and biochemical analysis of cerebral tissue. KEY RESULTS Doxy treatment abolished cognitive and daily life activity deficiencies in A53T mice. The effect on cognitive functions was associated with neuroprotection, inhibition of α-syn oligomerization and gliosis both in the cortex and hippocampus. Doxy treatment restored hippocampal long-term potentiation in association with the inhibition of pro-inflammatory cytokines expression. Moreover, Doxy ameliorated motor impairment and reduced striatal glial activation in A53T mice. CONCLUSIONS AND IMPLICATIONS Our findings promote Doxy as a valuable multi-target therapeutic approach counteracting both symptoms and neuropathology in the complex scenario of α-synucleinopathies.
Collapse
|
16
|
Ayeni EA, Aldossary AM, Ayejoto DA, Gbadegesin LA, Alshehri AA, Alfassam HA, Afewerky HK, Almughem FA, Bello SM, Tawfik EA. Neurodegenerative Diseases: Implications of Environmental and Climatic Influences on Neurotransmitters and Neuronal Hormones Activities. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph191912495. [PMID: 36231792 PMCID: PMC9564880 DOI: 10.3390/ijerph191912495] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 05/23/2023]
Abstract
Neurodegenerative and neuronal-related diseases are major public health concerns. Human vulnerability to neurodegenerative diseases (NDDs) increases with age. Neuronal hormones and neurotransmitters are major determinant factors regulating brain structure and functions. The implications of environmental and climatic changes emerged recently as influence factors on numerous diseases. However, the complex interaction of neurotransmitters and neuronal hormones and their depletion under environmental and climatic influences on NDDs are not well established in the literature. In this review, we aim to explore the connection between the environmental and climatic factors to NDDs and to highlight the available and potential therapeutic interventions that could use to improve the quality of life and reduce susceptibility to NDDs.
Collapse
Affiliation(s)
- Emmanuel A. Ayeni
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ahmad M. Aldossary
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Daniel A. Ayejoto
- Department of Industrial Chemistry, University of Ilorin, Ilorin 240003, Nigeria
| | - Lanre A. Gbadegesin
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Mountain Hazards and Environment, Chinese Academy of Sciences, Chengdu 610041, China
| | - Abdullah A. Alshehri
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Haya A. Alfassam
- KACST-BWH Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Henok K. Afewerky
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- School of Allied Health Professions, Asmara College of Health Sciences, Asmara P.O. Box 1220, Eritrea
| | - Fahad A. Almughem
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Saidu M. Bello
- Institute of Pharmacognosy, University of Szeged, 6720 Szeged, Hungary
| | - Essam A. Tawfik
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| |
Collapse
|
17
|
Lee DW, Ryu YK, Chang DH, Park HY, Go J, Maeng SY, Hwang DY, Kim BC, Lee CH, Kim KS. Agathobaculum butyriciproducens Shows Neuroprotective Effects in a 6-OHDA-Induced Mouse Model of Parkinson's Disease. J Microbiol Biotechnol 2022; 32:1168-1177. [PMID: 36168204 PMCID: PMC9628974 DOI: 10.4014/jmb.2205.05032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is the second-most prevalent neurodegenerative disease and is characterized by dopaminergic neuronal death in the midbrain. Recently, the association between alterations in PD pathology and the gut microbiota has been explored. Microbiota-targeted interventions have been suggested as a novel therapeutic approach for PD. Agathobaculum butyriciproducens SR79T (SR79) is an anaerobic bacterium. Previously, we showed that SR79 treatment induced cognitive improvement and reduced Alzheimer's disease pathologies in a mouse model. In this study, we hypothesized that SR79 treatment may have beneficial effects on PD pathology. To investigate the therapeutic effects of SR79 on PD, 6-hydroxydopamine (6-OHDA)-induced mouse models were used. D-Amphetamine sulfate (d-AMPH)-induced behavioral rotations and dopaminergic cell death were analyzed in unilateral 6-OHDA-lesioned mice. Treatment with SR79 significantly decreased ipsilateral rotations induced by d-AMPH. Moreover, SR79 treatment markedly activated the AKT/GSK3β signaling pathway in the striatum. In addition, SR79 treatment affected the Nrf2/ARE signaling pathway and its downstream target genes in the striatum of 6-OHDA-lesioned mice. Our findings suggest a protective role of SR79 in 6-OHDA-induced toxicity by regulating the AKT/Nrf2/ARE signaling pathway and astrocyte activation. Thus, SR79 may be a potential microbe-based intervention and therapeutic strategy for PD.
Collapse
Affiliation(s)
- Da Woon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,Department of Biomaterials Science, College of Natural Resources and Life Science and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Young-Kyoung Ryu
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Dong-Ho Chang
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Hye-Yeon Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Jun Go
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - So-Young Maeng
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,College of Biosciences and Biotechnology, Chung-Nam National University, Daejeon 34134, Republic of Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science, College of Natural Resources and Life Science and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Byoung-Chan Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,HealthBiome, Inc., Daejeon 34141, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,Department of Biosystems and Bioengineering, University of Science and Technology (UST), Daejeon 34113, Republic of Korea,Corresponding authors C.H. Lee E-mail:
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,
K.S. Kim Phone: 82-42-860-4634 Fax : 82-42-860-4609 E-mail:
| |
Collapse
|
18
|
Ushio S, Wada Y, Nakamura M, Matsumoto D, Hoshika K, Shiromizu S, Iwata N, Esumi S, Kajizono M, Kitamura Y, Sendo T. Anxiolytic-like effects of hochuekkito in lipopolysaccharide-treated mice involve interleukin-6 inhibition. Front Pharmacol 2022; 13:890048. [PMID: 36034871 PMCID: PMC9411515 DOI: 10.3389/fphar.2022.890048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Hochuekkito (HET) is a Kampo medicine used to treat postoperative and post-illness general malaise and decreased motivation. HET is known to regulate immunity and modulate inflammation. However, the precise mechanism and effects of HET on inflammation-induced central nervous system disorders remain unclear. This study aimed to assess the effect of HET on inflammation-induced anxiety-like behavior and the mechanism underlying anxiety-like behavior induced by lipopolysaccharide (LPS). Institute of Cancer Research mice were treated with LPS (300 μg/kg, intraperitoneally), a bacterial endotoxin, to induce systemic inflammation. The mice were administered HET (1.0 g/kg, orally) once a day for 2 weeks before LPS treatment. The light-dark box test and the hole-board test were performed 24 h after the LPS injection to evaluate the effects of HET on anxiety-like behaviors. Serum samples were obtained at 2, 5, and 24 h after LPS injection, and interleukin-6 (IL-6) levels in serum were measured. Human and mouse macrophage cells (THP-1 and RAW264.7 cells, respectively) were used to investigate the effect of HET on LPS-induced IL-6 secretion. The repeated administration of HET prevented anxiety-like behavior and decreased serum IL-6 levels in LPS-treated mice. HET significantly suppressed LPS-induced IL-6 secretion in RAW264.7 and THP-1 cells. Similarly, glycyrrhizin, one of the chemical constituents of HET, suppressed LPS-induced anxiety-like behaviors. Our study revealed that HET ameliorated LPS-induced anxiety-like behavior and inhibited IL-6 release in vivo and in vitro. Therefore, we postulate that HET may be useful against inflammation-induced anxiety-like behavior.
Collapse
Affiliation(s)
- Soichiro Ushio
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Yudai Wada
- Department of Clinical Pharmacy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mizuki Nakamura
- Department of Clinical Pharmacy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Daiki Matsumoto
- Department of Clinical Pharmacy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kota Hoshika
- Department of Clinical Pharmacy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shoya Shiromizu
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Naohiro Iwata
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Satoru Esumi
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Makoto Kajizono
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Yoshihisa Kitamura
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
- Department of Pharmacotherapy, School of Pharmacy, Shujitsu University, Okayama, Japan
- *Correspondence: Yoshihisa Kitamura,
| | - Toshiaki Sendo
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
19
|
Ivan I, Irincu L, Diaconu Ş, Falup-Pecurariu C. Parkinsonism associated with viral infection. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 165:1-16. [PMID: 36208896 DOI: 10.1016/bs.irn.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
There are several known causes of secondary parkinsonism, the most common being head trauma, stroke, medications, or infections. A growing body of evidence suggests that viral agents may trigger parkinsonian symptoms, but the exact pathological mechanisms are still unknown. In some cases, lesions or inflammatory processes in the basal ganglia or substantia nigra have been found to cause reversible or permanent impairment of the dopaminergic pathway, leading to the occurrence of extrapyramidal symptoms. This chapter reviews current data regarding the viral agents commonly associated with parkinsonism, such as Epstein Barr virus (EBV), hepatitis viruses, human immunodeficiency virus (HIV), herpes viruses, influenza virus, coxsackie virus, and Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). We present possible risk factors, proposed pathophysiology mechanisms, published case reports, common associations, and prognosis in order to offer a concise overview of the viral spectrum involved in parkinsonism.
Collapse
Affiliation(s)
| | | | - Ştefania Diaconu
- County Clinic Hospital, Brașov, Romania; Faculty of Medicine, Transilvania University, Brașov, Romania.
| | - Cristian Falup-Pecurariu
- County Clinic Hospital, Brașov, Romania; Faculty of Medicine, Transilvania University, Brașov, Romania
| |
Collapse
|
20
|
Mamais A, Kaganovich A, Harvey K. Convergence of signalling pathways in innate immune responses and genetic forms of Parkinson's disease. Neurobiol Dis 2022; 169:105721. [PMID: 35405260 DOI: 10.1016/j.nbd.2022.105721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 10/18/2022] Open
Abstract
In recent years progress in molecular biology and genetics have advanced our understanding of neurological disorders and highlighted synergistic relationships with inflammatory and age-related processes. Parkinson's disease (PD) is a common neurodegenerative disorder that is characterized by loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Increasing extensive evidence supports the contribution of genetic risk variants and inflammation in the pathobiology of this disease. Functional and genetic studies demonstrate an overlap between genes linked to increased risk for PD and autoimmune diseases. Variants identified in loci adjacent to LRRK2, GBA, and HLA establish a crosstalk between the pathobiologies of the two disease spectra. Furthermore, common signalling pathways associated with the pathogenesis of genetic PD are also relevant to inflammatory signaling include MAPK, NF-κB, Wnt and inflammasome signaling. Importantly, post-mortem analyses of brain and cerebrospinal fluid from PD patients show the accumulation of proinflammatory cytokines. In this review we will focus on the principal mechanisms of genetic, inflammatory and age-related risk that intersect in the pathogenesis of PD.
Collapse
Affiliation(s)
- Adamantios Mamais
- Department of Neurology, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Alice Kaganovich
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK..
| |
Collapse
|
21
|
Fang J, She J, Lin F, Wu JC, Han R, Sheng R, Wang G, Qin ZH. RRx-001 Exerts Neuroprotection Against LPS-Induced Microglia Activation and Neuroinflammation Through Disturbing the TLR4 Pathway. Front Pharmacol 2022; 13:889383. [PMID: 35462935 PMCID: PMC9020799 DOI: 10.3389/fphar.2022.889383] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/22/2022] [Indexed: 12/17/2022] Open
Abstract
Neuroinflammation plays an important role in the pathogenesis of many central nervous system diseases. Here, we investigated the effect of an anti-cancer compound RRx-001 on neuroinflammation and its possible new applications. BV2 cells and primary microglia cells were used to evaluate the role of RRx-001 in LPS-induced microglial activation and inflammatory response in vitro. And, we found that the increase in the synthesis and release of cytokines and the up-regulation of pro-inflammatory factors in LPS-treated microglial cells were significantly reduced by RRx-001 pretreatment. As the most classical inflammatory pathways, NF-κB and MAPK signaling pathways were activated by LPS, but were inhibited by RRx-001. Transcription of NLRP3 was also reduced by RRx-001. In addition, LPS induced oxidative stress by increasing the expression of Nox mediated by transcription factors NF-κB and AP-1, while RRx-001 pretreatment ameliorated Nox-mediated oxidative stress. LPS-induced activation of TAK1, an upstream regulator of NF-κB and MAPK pathways, was significantly inhibited by RRx-001 pretreatment, whereas recruitment of MyD88 to TLR4 was not affected by RRx-001. LPS-primed BV2 condition medium induced injury of primary neurons, and this effect was inhibited by RRx-001. Furthermore, we established a neuroinflammatory mouse model by stereotactic injection of LPS into the substantia nigra pars compacta (SNpc), and RRx-001 dose-dependently reduced LPS-induced microglial activation and loss of TH + neurons in the midbrain. In conclusion, the current study found that RRx-001 suppressed microglia activation and neuroinflammation through targeting TAK1, and may be a candidate for the treatment of neuroinflammation-related brain diseases.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jing She
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Fang Lin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jun-Chao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Guanghui Wang
- Department of Pharmacology and Laboratory of Molecular Pathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
22
|
Josiah SS, Famusiwa CD, Crown OO, Lawal AO, Olaleye MT, Akindahunsi AA, Akinmoladun AC. Neuroprotective effects of catechin and quercetin in experimental Parkinsonism through modulation of dopamine metabolism and expression of IL-1β, TNF-α, NF-κB, IκKB, and p53 genes in male Wistar rats. Neurotoxicology 2022; 90:158-171. [PMID: 35337893 DOI: 10.1016/j.neuro.2022.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
The neurobehavioral, brain redox-stabilizing and neurochemical modulatory properties of catechin and quercetin in rotenone-induced Parkinsonism, and the involvement of NF-κB-mediated inflammation, were investigated. Male Wistar rats subcutaneously administered with multiple doses of 1.5mg/kg rotenone were post-treated with 5-20mg/kg catechin or quercetin. This was followed by neurobehavioral evaluation, biochemical estimations, and assessment of neurotransmitter metabolism in the striatum. Expression of genes involved in the canonical pathway for the activation of NF-κB mediated inflammation (IL-1β, TNF-α, NF-κB, and IκKB) and the pro-apoptotic gene, p53, in the striatum was determined by RT-qPCR. Catechin and quercetin mitigated neurobehavioral deficits caused by rotenone. Both flavonoids attenuated striatal redox stress and neurochemical dysfunction, optimized disturbed dopamine metabolism, and improved depletion of neuron density caused by rotenone toxicity. While administration of catechin produced a more pronounced attenuating effect on IL-1β, TNF-α, and p53 genes, the attenuating effect of quercetin (20mg/kg) was more pronounced on NF-κB and IκKB gene expressions when compared to the group administered with rotenone only. Comparatively, quercetin demonstrated superior protection against rotenone neurotoxicity. It is concluded that catechin and quercetin have potential relevance in Parkinson's disease therapy through amelioration of redox stress, optimization of dopamine metabolism, and modulation of anti-inflammatory and anti-apoptotic pathways.
Collapse
Affiliation(s)
- Sunday Solomon Josiah
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria; Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - Courage Dele Famusiwa
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria; Department of Chemical sciences, Skyline University Nigeria, Kano, Nigeria
| | - Olamide Olajusi Crown
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria; Department of Chemistry, Physics and Atmospheric Science, Jackson State University, Jackson, MS 39204, USA
| | - Akeem O Lawal
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| | - Mary Tolulope Olaleye
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| | - Afolabi Akintunde Akindahunsi
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| | - Afolabi Clement Akinmoladun
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria.
| |
Collapse
|
23
|
Usama Ashhar M, Vyas P, Vohora D, Kumar Sahoo P, Nigam K, Dang S, Ali J, Baboota S. Amelioration of oxidative stress utilizing nanoemulsion loaded with bromocriptine and glutathione for the management of Parkinson's disease. Int J Pharm 2022; 618:121683. [PMID: 35314276 DOI: 10.1016/j.ijpharm.2022.121683] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 10/18/2022]
Abstract
Parkinson's disease (PD) is triggered by the formation of free radicals in dopaminergic neurons, which results in oxidative stress-induced neurodegeneration. The objective of the work was to relieve oxidative stress by employing intranasal delivery of Bromocriptine Mesylate (BRM) and Glutathione (GSH) loaded nanoemulsion for the better management of PD. The depth of permeation of the nanoemulsion was assessed through confocal laser scanning microscopy (CLSM) which revealed higher nanoemulsion permeation in contrast to suspension. Biocompatibility of nanoemulsion was confirmed by nasal cilio toxicity study. The DPPH study showed that the nanoemulsion had significant antioxidant activity. Biochemical estimation studies in Wistar rats were carried out in order to determine the effect of nanoemulsion on oxidative stress. The levels of GSH, superoxide dismutase (SOD), and catalase (CAT) were significantly enhanced; and the level of thiobarbituric acid reactive substances (TBARS) was significantly reduced after the intranasal administration of nanoemulsion in the haloperidol-induced model of PD. Furthermore, the levels of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) were also determined which reduced significantly after the administration of nanoemulsion. The oxidative stress levels were lowered with nanoemulsion, showing the combined antioxidant capability of BRM and GSH. The neuroprotective effect of the prepared nanoemulsion was confirmed by histopathological studies. Pharmacokinetic study revealed a higher concentration of BRM and GSH in the brain of Wistar rats after intranasal administration of nanoemulsion with a higher Brain/Plasma ratio. A higher value of AUC(0-8) of nanoemulsion in the brain after intranasal administration revealed that BRM and GSH remained in the brain for a longer period due to sustained release from nanoemulsion. According to the findings, BRM and GSH loaded nanoemulsion has the potential to provide a combined and synergistic anti-oxidant effect for efficient management of PD.
Collapse
Affiliation(s)
- Muhammad Usama Ashhar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Preeti Vyas
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Pravat Kumar Sahoo
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Kuldeep Nigam
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh 201309, India
| | - Shweta Dang
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh 201309, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
24
|
The Protective Effect of a Unique Mix of Polyphenols and Micronutrients against Neurodegeneration Induced by an In Vitro Model of Parkinson’s Disease. Int J Mol Sci 2022; 23:ijms23063110. [PMID: 35328530 PMCID: PMC8955775 DOI: 10.3390/ijms23063110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 12/04/2022] Open
Abstract
Parkinson’s disease (PD) is second-most common disabling neurological disorder worldwide, and unfortunately, there is not yet a definitive way to prevent it. Polyphenols have been widely shown protective efficacy against various PD symptoms. However, data on their effect on physio-pathological mechanisms underlying this disease are still lacking. In the present work, we evaluated the activity of a mixture of polyphenols and micronutrients, named A5+, in the murine neuroblastoma cell line N1E115 treated with 6-Hydroxydopamine (6-OHDA), an established neurotoxic stimulus used to induce an in vitro PD model. We demonstrate that a pretreatment of these cells with A5+ causes significant reduction of inflammation, resulting in a decrease in pro-inflammatory cytokines (IFN-γ, IL-6, TNF-α, and CXCL1), a reduction in ROS production and activation of extracellular signal-regulated kinases (ERK)1/2, and a decrease in apoptotic mechanisms with the related increase in cell viability. Intriguingly, A5+ treatment promoted cellular differentiation into dopaminergic neurons, as evident by the enhancement in the expression of tyrosine hydroxylase, a well-established dopaminergic neuronal marker. Overall, these results demonstrate the synergic and innovative efficacy of A5+ mixture against PD cellular pathological processes, although further studies are needed to clarify the mechanisms underlying its beneficial effect.
Collapse
|
25
|
Bian M, Chen L, Lei L. Research progress on the relationship between chronic periodontitis and Parkinson's disease. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:108-114. [PMID: 35462470 PMCID: PMC9109767 DOI: 10.3724/zdxbyxb-2021-0111] [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: 10/24/2021] [Accepted: 01/25/2022] [Indexed: 06/14/2023]
Abstract
Chronic periodontitis is an infectious disease, which has a reciprocal relationship with a variety of systemic disorders. Parkinson's disease is a prevalent neurodegenerative disease in which inflammation plays an important role for its progression. A vast number of studies suggest that there is a potential connection between chronic periodontitis and neurodegenerative diseases such as Parkinson's disease. Individuals with Parkinson's disease usually have poor periodontal health, and their oral flora composition differs from that of healthy people; at the same time, patients with chronic periodontitis have a higher risk of Parkinson's disease, which can be reduced with regular periodontal treatment. In fact, the mechanism of interaction between chronic periodontitis and Parkinson's disease is not clear. According to several studies, the clinical symptoms of Parkinson's disease prevent patients to maintain oral hygiene effectively, increasing the risk of periodontitis. Neuroinflammation mediated by microglia may be the key to the influence of chronic periodontitis on Parkinson's disease. Periodontal pathogens and inflammatory mediators may enter the brain and activate microglia in various ways, and ultimately leading to occurrence and development of Parkinson's disease. This article reviews the recent research progress on the association between chronic periodontitis and Parkinson's disease, and its potential mechanism to provide information for further research.
Collapse
|
26
|
Akinmoladun AC, Famusiwa CD, Josiah SS, Lawal AO, Olaleye MT, Akindahunsi AA. Dihydroquercetin improves rotenone‐induced Parkinsonism by regulating NF‐κB‐mediated inflammation pathway in rats. J Biochem Mol Toxicol 2022; 36:e23022. [DOI: 10.1002/jbt.23022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/31/2021] [Accepted: 01/28/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Afolabi C. Akinmoladun
- Department of Biochemistry, School of Life Sciences The Federal University of Technology Akure Nigeria
| | - Courage D. Famusiwa
- Department of Biochemistry, School of Life Sciences The Federal University of Technology Akure Nigeria
| | - Sunday S. Josiah
- Department of Biochemistry, School of Life Sciences The Federal University of Technology Akure Nigeria
| | - Akeem O. Lawal
- Department of Biochemistry, School of Life Sciences The Federal University of Technology Akure Nigeria
| | - Mary T. Olaleye
- Department of Biochemistry, School of Life Sciences The Federal University of Technology Akure Nigeria
| | - Afolabi A. Akindahunsi
- Department of Biochemistry, School of Life Sciences The Federal University of Technology Akure Nigeria
| |
Collapse
|
27
|
Huntula S, Lalert L, Punsawad C. The Effects of Exercise on Aging-Induced Exaggerated Cytokine Responses: An Interdisciplinary Discussion. SCIENTIFICA 2022; 2022:3619362. [PMID: 35106183 PMCID: PMC8801319 DOI: 10.1155/2022/3619362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
Aging is generally known to be associated with dynamic biological changes, physiological dysfunction, and environmental and psychological decline. Several studies have suggested that aging is associated with increased inflammatory cytokines, causing several diseases. However, the effect of exercise on aging has been less delineated, and the relationships between cytokine activation, aging, and exercise also need further study. Here, we discuss some ideas about the effect of exercise on aging-induced exaggerated cytokine responses and discuss the possible roles of the aging-induced exaggerated cytokine response following exercise. Evidence from these findings suggests that exercise is a beneficially applicable model to use in studies on the mechanisms underlying the age-associated gradated cytokine response, and these results may provide guidelines for health professionals with diverse backgrounds.
Collapse
Affiliation(s)
- Soontaraporn Huntula
- Department of Sport and Exercise Science, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Laddawan Lalert
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Chuchard Punsawad
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
28
|
Morales-Rosales SL, Santín-Márquez R, Posadas-Rodriguez P, Rincon-Heredia R, Montiel T, Librado-Osorio R, Luna-López A, Rivero-Segura NA, Torres C, Cano-Martínez A, Silva-Palacios A, Cortés-Hernández P, Morán J, Massieu L, Konigsberg M. Senescence in Primary Rat Astrocytes Induces Loss of the Mitochondrial Membrane Potential and Alters Mitochondrial Dynamics in Cortical Neurons. Front Aging Neurosci 2021; 13:766306. [PMID: 34924995 PMCID: PMC8672143 DOI: 10.3389/fnagi.2021.766306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/27/2021] [Indexed: 01/10/2023] Open
Abstract
The decline in brain function during aging is one of the most critical health problems nowadays. Although senescent astrocytes have been found in old-age brains and neurodegenerative diseases, their impact on the function of other cerebral cell types is unknown. The aim of this study was to evaluate the effect of senescent astrocytes on the mitochondrial function of a neuron. In order to evaluate neuronal susceptibility to a long and constant senescence-associated secretory phenotype (SASP) exposure, we developed a model by using cellular cocultures in transwell plates. Rat primary cortical astrocytes were seeded in transwell inserts and induced to premature senescence with hydrogen peroxide [stress-induced premature senescence (SIPS)]. Independently, primary rat cortical neurons were seeded at the bottom of transwells. After neuronal 6 days in vitro (DIV), the inserts with SIPS-astrocytes were placed in the chamber and cocultured with neurons for 6 more days. The neuronal viability, the redox state [reduced glutathione/oxidized glutathione (GSH/GSSG)], the mitochondrial morphology, and the proteins and membrane potential were determined. Our results showed that the neuronal mitochondria functionality was altered after being cocultured with senescent astrocytes. In vivo, we found that old animals had diminished mitochondrial oxidative phosphorylation (OXPHOS) proteins, redox state, and senescence markers as compared to young rats, suggesting effects of the senescent astrocytes similar to the ones we observed in vitro. Overall, these results indicate that the microenvironment generated by senescent astrocytes can affect neuronal mitochondria and physiology.
Collapse
Affiliation(s)
- Sandra Lizbeth Morales-Rosales
- Posgrado Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Roberto Santín-Márquez
- Posgrado Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Pedro Posadas-Rodriguez
- Posgrado Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Ruth Rincon-Heredia
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Teresa Montiel
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Raúl Librado-Osorio
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Mexico City, Mexico
| | - Armando Luna-López
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Mexico City, Mexico
| | | | - Claudio Torres
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Paulina Cortés-Hernández
- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Oriente, Atlixco, Mexico
| | - Julio Morán
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lourdes Massieu
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mina Konigsberg
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| |
Collapse
|
29
|
Nebrisi EE. Neuroprotective Activities of Curcumin in Parkinson's Disease: A Review of the Literature. Int J Mol Sci 2021; 22:11248. [PMID: 34681908 PMCID: PMC8537234 DOI: 10.3390/ijms222011248] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 08/30/2021] [Accepted: 09/04/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is a slowly progressive multisystem disorder affecting dopaminergic neurons of the substantia nigra pars compacta (SNpc), which is characterized by a decrease of dopamine (DA) in their striatal terminals. Treatment of PD with levodopa or DA receptor agonists replaces the function of depleted DA in the striatum. Prolonged treatment with these agents often has variable therapeutic effects and leads to the development of undesirable dyskinesia. Consequently, a crucial unmet demand in the management of Parkinson's disease is the discovery of new approaches that could slow down, stop, or reverse the process of neurodegeneration. Novel potential treatments involving natural substances with neuroprotective activities are being developed. Curcumin is a polyphenolic compound isolated from the rhizomes of Curcuma longa (turmeric). It has been demonstrated to have potent anti-inflammatory, antioxidant, free radical scavenging, mitochondrial protecting, and iron-chelating effects, and is considered a promising therapeutic and nutraceutical agent for the treatment of PD. However, molecular and cellular mechanisms that mediate the pharmacological actions of curcumin remain largely unknown. Stimulation of nicotinic receptors and, more precisely, selective α7 nicotinic acetylcholine receptors (α7-nAChR), have been found to play a major modulatory role in the immune system via the "cholinergic anti-inflammatory pathway". Recently, α7-nAChR has been proposed to be a potential therapeutic approach in PD. In this review, the detailed mechanisms of the neuroprotective activities of curcumin as a potential therapeutic agent to help Parkinson's patients are being discussed and elaborated on in detail.
Collapse
Affiliation(s)
- Eslam El Nebrisi
- Department of Pharmacology, Dubai Medical College, Dubai 20170, United Arab Emirates
| |
Collapse
|
30
|
Sanchez A, Morales I, Rodriguez-Sabate C, Sole-Sabater M, Rodriguez M. Astrocytes, a Promising Opportunity to Control the Progress of Parkinson's Disease. Biomedicines 2021; 9:biomedicines9101341. [PMID: 34680458 PMCID: PMC8533570 DOI: 10.3390/biomedicines9101341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 12/21/2022] Open
Abstract
At present, there is no efficient treatment to prevent the evolution of Parkinson’s disease (PD). PD is generated by the concurrent activity of multiple factors, which is a serious obstacle for the development of etio-pathogenic treatments. Astrocytes may act on most factors involved in PD and the promotion of their neuroprotection activity may be particularly suitable to prevent the onset and progression of this basal ganglia (BG) disorder. The main causes proposed for PD, the ability of astrocytes to control these causes, and the procedures that can be used to promote the neuroprotective action of astrocytes will be commented upon, here.
Collapse
Affiliation(s)
- Alberto Sanchez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain; (A.S.); (I.M.); (C.R.-S.)
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Ingrid Morales
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain; (A.S.); (I.M.); (C.R.-S.)
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Clara Rodriguez-Sabate
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain; (A.S.); (I.M.); (C.R.-S.)
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Department of Psychiatry, Getafe University Hospital, 28905 Madrid, Spain
| | - Miguel Sole-Sabater
- Department of Neurology, La Candelaria University Hospital, 38010 Tenerife, Spain;
| | - Manuel Rodriguez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain; (A.S.); (I.M.); (C.R.-S.)
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Correspondence: ; Tel.: +34-922-319361; Fax: +34-922-319397
| |
Collapse
|
31
|
Guglietti B, Sivasankar S, Mustafa S, Corrigan F, Collins-Praino LE. Fyn Kinase Activity and Its Role in Neurodegenerative Disease Pathology: a Potential Universal Target? Mol Neurobiol 2021; 58:5986-6005. [PMID: 34432266 DOI: 10.1007/s12035-021-02518-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Fyn is a non-receptor tyrosine kinase belonging to the Src family of kinases (SFKs) which has been implicated in several integral functions throughout the central nervous system (CNS), including myelination and synaptic transmission. More recently, Fyn dysfunction has been associated with pathological processes observed in neurodegenerative diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD) and Parkinson's disease (PD). Neurodegenerative diseases are amongst the leading cause of death and disability worldwide and, due to the ageing population, prevalence is predicted to rise in the coming years. Symptoms across neurodegenerative diseases are both debilitating and degenerative in nature and, concerningly, there are currently no disease-modifying therapies to prevent their progression. As such, it is important to identify potential new therapeutic targets. This review will outline the role of Fyn in normal/homeostatic processes, as well as degenerative/pathological mechanisms associated with neurodegenerative diseases, such as demyelination, pathological protein aggregation, neuroinflammation and cognitive dysfunction.
Collapse
Affiliation(s)
- Bianca Guglietti
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Srisankavi Sivasankar
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Sanam Mustafa
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia
| | - Frances Corrigan
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia
| | - Lyndsey E Collins-Praino
- Department of Medical Sciences, University of Adelaide, SG31, Helen Mayo South, Adelaide, SA, 5005, Australia. .,ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
32
|
Tiefensee Ribeiro C, Peixoto DO, Santos L, Saibro-Girardi C, Brum PO, Carazza-Kessler FG, Somensi N, Behrens LMP, Bittencourt RR, Soares LS, Silveira AK, de Oliveira J, Moreira JCF, Gasparotto J, Gelain DP. Intranasal HSP70 administration protects against dopaminergic denervation and modulates neuroinflammatory response in the 6-OHDA rat model. Brain Behav Immun Health 2021; 14:100253. [PMID: 34589762 PMCID: PMC8474599 DOI: 10.1016/j.bbih.2021.100253] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/22/2021] [Accepted: 03/28/2021] [Indexed: 11/17/2022] Open
Abstract
HSP70 is one of the main molecular chaperones involved in the cellular stress response. Besides its chaperone action, HSP70 also modulates the immune response. Increased susceptibility to toxic insults in intra- and extracellular environments has been associated with insufficient amounts of inducible HSP70 in adult neurons. On the other hand, exogenous HSP70 administration has demonstrated neuroprotective effects in experimental models of age-related disorders. In this regard, this study investigated the effects of exogenous HSP70 in an animal model of dopaminergic denervation of the nigrostriatal axis. After unilateral intrastriatal injection with 6-hydroxydopamine (6-OHDA), the animals received purified recombinant HSP70 through intranasal administration (2 μg/rat/day) for 15 days. Our results indicate a neuroprotective effect of intranasal HSP70 against dopaminergic denervation induced by 6-OHDA. Exogenous HSP70 improved motor impairment and reduced the loss of dopaminergic neurons caused by 6-OHDA. Moreover, HSP70 modulated neuroinflammatory response in the substantia nigra, an important event in Parkinson's disease pathogenesis. Specifically, HSP70 treatment reduced microglial activation and astrogliosis induced by 6-OHDA, as well as IL-1β mRNA expression in this region. Also, recombinant HSP70 increased the protein content of HSP70 in the substantia nigra of rats that received 6-OHDA. These data suggest the neuroprotection of HSP70 against dopaminergic neurons damage after cellular stress. Finally, our results indicate that HSP70 neuroprotective action against 6-OHDA toxicity is related to inflammatory response modulation.
Collapse
Affiliation(s)
- Camila Tiefensee Ribeiro
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daniel Oppermann Peixoto
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lucas Santos
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carolina Saibro-Girardi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pedro Ozorio Brum
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Flávio Gabriel Carazza-Kessler
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Nauana Somensi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luiza Marques Prates Behrens
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Reykla Ramon Bittencourt
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Laíssa Santos Soares
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alexandre Kleber Silveira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Jade de Oliveira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - José Cláudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Juciano Gasparotto
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daniel Pens Gelain
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
33
|
Kwon O, Song J, Yang Y, Kim S, Kim JY, Seok M, Hwang I, Yu J, Karmacharya J, Maeng H, Kim J, Jho E, Ko SY, Son H, Chang M, Lee S. SGK1 inhibition in glia ameliorates pathologies and symptoms in Parkinson disease animal models. EMBO Mol Med 2021; 13:e13076. [PMID: 33646633 PMCID: PMC8033538 DOI: 10.15252/emmm.202013076] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 01/20/2021] [Accepted: 01/23/2021] [Indexed: 12/13/2022] Open
Abstract
Astrocytes and microglia are brain-resident glia that can establish harmful inflammatory environments in disease contexts and thereby contribute to the progression of neuronal loss in neurodegenerative disorders. Correcting the diseased properties of glia is therefore an appealing strategy for treating brain diseases. Previous studies have shown that serum/ glucocorticoid related kinase 1 (SGK1) is upregulated in the brains of patients with various neurodegenerative disorders, suggesting its involvement in the pathogenesis of those diseases. In this study, we show that inhibiting glial SGK1 corrects the pro-inflammatory properties of glia by suppressing the intracellular NFκB-, NLRP3-inflammasome-, and CGAS-STING-mediated inflammatory pathways. Furthermore, SGK1 inhibition potentiated glial activity to scavenge glutamate toxicity and prevented glial cell senescence and mitochondrial damage, which have recently been reported as critical pathologic features of and therapeutic targets in Parkinson disease (PD) and Alzheimer disease (AD). Along with those anti-inflammatory/neurotrophic functions, silencing and pharmacological inhibition of SGK1 protected midbrain dopamine neurons from degeneration and cured pathologic synuclein alpha (SNCA) aggregation and PD-associated behavioral deficits in multiple in vitro and in vivo PD models. Collectively, these findings suggest that SGK1 inhibition could be a useful strategy for treating PD and other neurodegenerative disorders that share the common pathology of glia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Oh‐Chan Kwon
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Jae‐Jin Song
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
| | - Yunseon Yang
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Seong‐Hoon Kim
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Ji Young Kim
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Min‐Jong Seok
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Inhwa Hwang
- Korea Department of Microbiology and ImmunologyInstitute for Immunology and Immunological DiseasesBrain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulSouth Korea
| | - Je‐Wook Yu
- Korea Department of Microbiology and ImmunologyInstitute for Immunology and Immunological DiseasesBrain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulSouth Korea
| | | | | | - Jiyoung Kim
- Department of Life ScienceUniversity of SeoulSeoulKorea
| | - Eek‐hoon Jho
- Department of Life ScienceUniversity of SeoulSeoulKorea
| | - Seung Yeon Ko
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Hyeon Son
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| | - Mi‐Yoon Chang
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
| | - Sang‐Hun Lee
- Department of Biochemistry and Molecular BiologyCollege of MedicineHanyang UniversitySeoulKorea
- Hanyang Biomedical Research InstituteHanyang UniversitySeoulKorea
- Graduate School of Biomedical Science and EngineeringHanyang UniversitySeoul
| |
Collapse
|
34
|
Silva J, Alves C, Pinteus S, Susano P, Simões M, Guedes M, Martins A, Rehfeldt S, Gaspar H, Goettert M, Alfonso A, Pedrosa R. Disclosing the potential of eleganolone for Parkinson's disease therapeutics: Neuroprotective and anti-inflammatory activities. Pharmacol Res 2021; 168:105589. [PMID: 33812007 DOI: 10.1016/j.phrs.2021.105589] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022]
Abstract
The treatment of Parkinson´s disease (PD) has benefited from significant advances resulting from the increasing research efforts focused on new therapeutics. However, the current treatments for PD are mostly symptomatic, alleviating disease symptoms without reversing or retarding disease progression. Thus, it is critical to find new molecules that can result in more effective treatments. Within this framework, this study aims to evaluate the neuroprotective and anti-inflammatory effects of three compounds (eleganolone, eleganonal and fucosterol) isolated from the brown seaweed Bifurcaria bifurcata. In vitro neuroprotective effects were evaluated on a PD cellular model induced by the neurotoxin 6-hydroxydopamine (6-OHDA) on SH-SY5Y human cells, while lipopolysaccharide (LPS) - stimulated RAW 264.7 macrophages were used to evaluate the anti-inflammatory potential. Additionally, the underlying mechanisms of action were also investigated. Compounds were isolated by preparative chromatographic methods and their structural elucidation attained by NMR spectroscopy. Among the tested compounds, eleganolone (0.1-1 µM; 24 h) reverted the neurotoxicity induced by 6-OHDA in about 20%. The neuroprotective effects were mediated by mitochondrial protection, reduction of oxidative stress, inflammation and apoptosis, and inhibition of NF-kB pathway. The results suggest that eleganolone may provide advantages in the treatment of neurodegenerative conditions and, therefore, should be considered for future preclinical studies.
Collapse
Affiliation(s)
- Joana Silva
- MARE - Marine and Environmental Sciences Centre, Polytechnic of Leiria, 2520-630 Peniche, Portugal; Department of Pharmacology, Faculty of Veterinary, University of Santiago de Compostela, 27002 Lugo, Spain.
| | - Celso Alves
- MARE - Marine and Environmental Sciences Centre, Polytechnic of Leiria, 2520-630 Peniche, Portugal
| | - Susete Pinteus
- MARE - Marine and Environmental Sciences Centre, Polytechnic of Leiria, 2520-630 Peniche, Portugal
| | - Patrícia Susano
- MARE - Marine and Environmental Sciences Centre, Polytechnic of Leiria, 2520-630 Peniche, Portugal
| | - Marco Simões
- MARE - Marine and Environmental Sciences Centre, Polytechnic of Leiria, 2520-630 Peniche, Portugal
| | - Miguel Guedes
- MARE - Marine and Environmental Sciences Centre, Polytechnic of Leiria, 2520-630 Peniche, Portugal
| | - Alice Martins
- MARE - Marine and Environmental Sciences Centre, Polytechnic of Leiria, 2520-630 Peniche, Portugal
| | - Stephanie Rehfeldt
- Cell Culture Laboratory, Graduate Program in Biotechnology, University of Vale do Taquari - UNIVATES, 95901-120 Lajeado, RS, Brazil
| | - Helena Gaspar
- MARE - Marine and Environmental Sciences Centre, Polytechnic of Leiria, 2520-630 Peniche, Portugal; BioISI - Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisboa, Portugal
| | - Márcia Goettert
- Cell Culture Laboratory, Graduate Program in Biotechnology, University of Vale do Taquari - UNIVATES, 95901-120 Lajeado, RS, Brazil
| | - Amparo Alfonso
- Department of Pharmacology, Faculty of Veterinary, University of Santiago de Compostela, 27002 Lugo, Spain
| | - Rui Pedrosa
- MARE - Marine and Environmental Sciences Centre, ESTM, Polytechnic of Leiria, 2520-614 Peniche, Portugal.
| |
Collapse
|
35
|
Zhao J, Kumar M, Sharma J, Yuan Z. Arbutin effectively ameliorates the symptoms of Parkinson's disease: the role of adenosine receptors and cyclic adenosine monophosphate. Neural Regen Res 2021; 16:2030-2040. [PMID: 33642391 PMCID: PMC8343309 DOI: 10.4103/1673-5374.308102] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
An antagonistic communication exists between adenosinergic and dopaminergic signaling in the basal ganglia, which suggests that the suppression of adenosine A2A receptors-cyclic adenosine monophosphate pathway may be able to restore the disrupted dopamine transmission that results in motor symptoms in Parkinson’s disease (PD). Arbutin is a natural glycoside that possesses antioxidant, anti-inflammatory, and neuroprotective properties. The purpose of this study was to investigate whether arbutin could ameliorate the symptoms of PD and to examine the underlying mechanism. In this study, Swiss albino mouse models of PD were established by the intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine for 4 successive days, with the concurrent intraperitoneal administration of arbutin (50 and 100 mg/kg) for 7 days. The results showed that arbutin significantly reduced lipid peroxidation, total nitrite levels, and inflammation in the substantia nigra and striatum of PD mouse models. In addition, arbutin decreased the activity of endogenous antioxidants, reduced the levels of dopamine, 3,4-dihydroxyphenylacetic acid, homovanillic acid, and γ-aminobutyric acid, and minimized neurodegeneration in the striatum. Arbutin also reduced the abnormal performance of PD mouse models in the open field test, bar test, pole test, and rotarod test. The therapeutic efficacy of arbutin was similar to that of madopar. The intraperitoneal injection of the A2AR agonist CGS21680 (0.5 mg/kg) attenuated the therapeutic effects of arbutin, whereas the intraperitoneal injection of forskolin (3 mg/kg) enhanced arbutin-mediated improvements. These findings suggest that arbutin can improve the performance of PD mouse models by inhibiting the function of the A2AR and enhancing the effects of cyclic adenosine monophosphate. This study was approved by the Institutional Animal Ethics Committee (1616/PO/Re/S/12/CPCSEA) on November 17, 2019 (approval No. IAEC/2019/010).
Collapse
Affiliation(s)
- Jie Zhao
- Department of Neurology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, China
| | - Manish Kumar
- Department of Pharmacology, Swift School of Pharmacy, Rajpura (Patiala), Punjab, India
| | - Jeevan Sharma
- Department of Pharmacology, Swift School of Pharmacy, Rajpura (Patiala), Punjab, India
| | - Zhihai Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
36
|
Parra-Paz VG, Calderón-Sauri A, Granados-Patrón D, Cuevas-Carbonell SG, García-López D, Dawn-Ojeda A, Mut-Martín M, Olivera-Castillo L, Álvarez-Cervera FJ, Salgado H, Alamilla J, García-Miss MDR, Vásquez-Celaya L, Aranda-González II, Góngora-Alfaro JL. Chronic feeding with 3% dried raw blueberries (V. corymbosum) reduces apomorphine-induced rotations and striatal dopaminergic loss in hemiparkinsonian rats. Food Res Int 2020; 140:110066. [PMID: 33648289 DOI: 10.1016/j.foodres.2020.110066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 12/09/2020] [Accepted: 12/20/2020] [Indexed: 11/28/2022]
Abstract
Blueberries (BB) are rich in antioxidant polyphenols, and their intake could prevent Parkinson's disease (PD). Here we assessed whether rats chronically fed dried raw BB develop resistance to dopaminergic denervation and motor disorders caused by unilateral intrastriatal injection of 6-hydroxydopamine (6-OHDA), a dopaminergic neurotoxin acting mainly by inducing oxidative stress. Male rats were fed either with LabDiet® alone or supplemented with 3% lyophilized raw BB for 2 weeks before and 3 weeks after injecting 6-OHDA (day 0) or vehicle (mock lesion) into the right striatum. The cylinder test was performed on days -14, -7, -1, +7, +14, and +21; the percentage of ipsilateral forepaw (IF) use asymmetry was determined by counting the wall contacts made with either forepaw or with both. Apomorphine (0.25 mg/kg, s.c.)-induced rotation was performed on days -1, +7, +14, and +21. Full contralateral rotations were counted in 3-min periods, every 15 min, up to 90 min. Striatal slices were immunostained for tyrosine hydroxylase (TH) and the ionized calcium-binding protein-1 adapter (Iba1) [immunoreactive area or microglia count in right striatum expressed as % of the left striatum]. Antioxidants in BB methanolic extracts neutralized the free radical 2,2-diphenyl-1-picrylhydrazyl in a concentration-dependent manner. Anthocyanins have been reported as the most abundant polyphenols in BB. Using the pH differential method, the total anthocyanin content (malvidin-3-glucoside equivalents) in raw BB averaged 21.04 mg/g dry weight. The range of anthocyanin intake by rats throughout the study varied from 37.7 to 72.2 mg/kg body weight. The time and food type factors, as well as their interaction were significant according to two-way RM-ANOVA in both the apomorphine-induced rotations and the cylinder test. Compared with LabDiet® alone, chronic supplementation with 3% dried raw BB decreased apomorphine-induced rotations on days +14 and +21 (p < 0.001) and produced a 46% reduction in total rotations post-surgery (p < 0.05), but only caused a partial, non-significant, decrease of IF asymmetry. BB supplementation reduced TH loss in the striatum (p < 0.05) but did not attenuate the increase of Iba1+ microglia. The consumption of 3% dried raw blueberries attenuates dopaminergic denervation and partially reverses motor disorders in the 6-OHDA-induced PD model in rats. The phytochemicals of raw blueberries that contribute to the observed neuroprotective effect are yet to be identified.
Collapse
Affiliation(s)
- Valeria G Parra-Paz
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida, Yucatán 97000, Mexico
| | - Ashanty Calderón-Sauri
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida, Yucatán 97000, Mexico
| | - Diego Granados-Patrón
- Facultad de Medicina, Universidad Autónoma de Yucatán, Avenida Itzáes No. 498 x 59 y 59A, Mérida, Yucatán 97000, Mexico
| | - Sergio G Cuevas-Carbonell
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida, Yucatán 97000, Mexico
| | - Daniel García-López
- Facultad de Medicina, Universidad Autónoma de Yucatán, Avenida Itzáes No. 498 x 59 y 59A, Mérida, Yucatán 97000, Mexico
| | - Alicia Dawn-Ojeda
- Facultad de Medicina, Universidad Autónoma de Yucatán, Avenida Itzáes No. 498 x 59 y 59A, Mérida, Yucatán 97000, Mexico
| | - Mirza Mut-Martín
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida, Yucatán 97000, Mexico
| | - Leticia Olivera-Castillo
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Unidad Mérida, Carretera Antigua Progreso Km. 6, Mérida, Yucatán 97310, Mexico
| | - Fernando J Álvarez-Cervera
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida, Yucatán 97000, Mexico
| | - Humberto Salgado
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida, Yucatán 97000, Mexico
| | - Javier Alamilla
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, Colima 28045, Mexico
| | - María Del R García-Miss
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida, Yucatán 97000, Mexico
| | - Lizbeth Vásquez-Celaya
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida, Yucatán 97000, Mexico
| | - Irma I Aranda-González
- Facultad de Medicina, Universidad Autónoma de Yucatán, Avenida Itzáes No. 498 x 59 y 59A, Mérida, Yucatán 97000, Mexico
| | - José L Góngora-Alfaro
- Departamento de Neurociencias, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida, Yucatán 97000, Mexico.
| |
Collapse
|
37
|
Potential anti-neuroinflammatory compounds from Australian plants - A review. Neurochem Int 2020; 142:104897. [PMID: 33186611 DOI: 10.1016/j.neuint.2020.104897] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 12/23/2022]
Abstract
Neuroinflammation is a complex response to brain injury involving the activation of glia, release of inflammatory mediators, such as cytokines and chemokines, and generation of reactive oxygen and nitrogen species. Even though it is considered an event secondary to neuronal death or dysfunction, neuro-inflammation comprises a majority of the non-neuronal contributors to the cause and progression of neurodegenerative diseases like Alzheimer's Disease (AD), Parkinson's Disease (PD), Multiple Sclerosis (MS), Chronic Traumatic Encephalopathy (CTE) and others. As a result of the lack of effectiveness of current treatments for neurodegenerative diseases, neuroinflammation has become a legitimate therapeutic target for drug discovery, leading to the study of various in vivo and in vitro models of neuroinflammation. Several molecules sourced from plants have displayed anti-inflammatory properties in the study of neurodegenerative diseases. A group of these anti-inflammatory compounds has been classified as cytokine-suppressive anti-inflammatory drugs (CSAIDs), which target the pro-inflammatory AP1 and nuclear factor-κB signaling pathways and inhibit the expression of many pro-inflammatory cytokines, such as interleukin IL-1, IL-6, TNF-α, or nitric oxide. Australian plants, thriving amid the driest inhabited continent of the world, are an untapped source of chemical diversity in the form of secondary metabolites. These compounds are produced in response to biotic and abiotic stresses that the plants are exposed to in the highly biodiverse environment. This review is an attempt to highlight anti-inflammatory compounds isolated from Australian plants.
Collapse
|
38
|
Neuroinflammatory responses in Parkinson's disease: relevance of Ibuprofen in therapeutics. Inflammopharmacology 2020; 29:5-14. [PMID: 33052479 DOI: 10.1007/s10787-020-00764-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) pathogenesis inevitably involves neuroinflammatory responses attained through contribution of both neuron and glial cells. Investigation done in both experimental models of PD and in samples of PD patients suggested the involvement of both central and peripheral inflammatory responses during PD pathogenesis. Such neuroinflammatory responses could be regulated by neuron-glia interaction which is one of the recently focused areas in the field of disease diagnosis, pathogenesis and therapeutics. Such aggravated neuroinflammatory responses during PD are very well associated with augmented levels of cyclooxygenase (COX). An increased expression of cyclooxygenase (COX) with a concomitant increase in the prostaglandin E2 (PGE2) levels has been observed during PD pathology. Ibuprofen is one of the non-steroidal anti-inflammatory drugs (NSAID) and clinically being used for PD patients. This review focuses on the neuroinflammatory responses during PD pathology as well as the effect of ibuprofen on various disease related signaling factors and mechanisms involving nitrosative stress, neurotransmission, neuronal communication and peroxisome proliferator-activated receptor-γ. Such mechanistic effect of ibuprofen has been mostly reported in experimental models of PD and clinical investigations are still required. Since oxidative neuronal death is one of the major neurodegenerative mechanisms in PD, the antioxidant capacity of ibuprofen along with its antidepressant effects have also been discussed. This review will direct the readers towards fulfilling the existing gaps in the mechanistic aspect of ibuprofen and enhance its clinical relevance in PD therapeutics and probably in other age-related neurodegenerative diseases.
Collapse
|
39
|
Huang D, Li Q, Wang Y, Liu Z, Wang Z, Li H, Wang J, Su J, Ma Y, Yu M, Fei J, Huang F. Brain-specific NRSF deficiency aggravates dopaminergic neurodegeneration and impairs neurogenesis in the MPTP mouse model of Parkinson's disease. Aging (Albany NY) 2020; 11:3280-3297. [PMID: 31147527 PMCID: PMC6555471 DOI: 10.18632/aging.101979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/12/2019] [Indexed: 12/13/2022]
Abstract
Degeneration of the dopaminergic neurons in the substantia nigra and the resultant dopamine depletion from the striatum are the hallmarks of Parkinson's disease (PD) and are responsible for the disease's cardinal motor symptoms. The transcriptional repressor Neuron-Restrictive Silencer Factor (NRSF), also known as RE1-Silencing Transcription Factor (REST), was originally identified as a negative regulator of neuron-specific genes in non-neuronal cells. Our previous study showed that mice deficient in neuronal NRSF/REST expression were more vulnerable to the noxious effects of the dopaminergic neurotoxin MPTP. Here, we found that brain-specific deletion of NRSF/REST led to more severe damages to the nigrostriatal pathway and long-lasting behavioral impairments in mice challenged with MPTP. Moreover, compared to wild-type controls, these mice showed increased neurogenesis shortly after MPTP exposure, but reduced neurogenesis later on. These results suggest that NRSF/REST acts as a negative modulator of neurogenesis and a pro-survival factor of neural stem cells under both normal conditions and during the course of PD.
Collapse
Affiliation(s)
- Dongping Huang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Qing Li
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yi Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhaolin Liu
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zishan Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Heng Li
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Jinghui Wang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Jing Su
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yuanyuan Ma
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Jian Fei
- School of Life Science and Technology, Tongji University, Shanghai 200092, China.,Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, Inc., Shanghai 201203, China
| | - Fang Huang
- Department of Translational Neuroscience, Jing' an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
40
|
Morales I, Sanchez A, Puertas-Avendaño R, Rodriguez-Sabate C, Perez-Barreto A, Rodriguez M. Neuroglial transmitophagy and Parkinson's disease. Glia 2020; 68:2277-2299. [PMID: 32415886 DOI: 10.1002/glia.23839] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 12/28/2022]
Abstract
Mitophagy is essential for the health of dopaminergic neurons because mitochondrial damage is a keystone of Parkinson's disease. The aim of the present work was to study the degradation of mitochondria in the degenerating dopaminergic synapse. Adult Sprague-Dawley rats and YFP-Mito-DAn mice with fluorescent mitochondria in dopaminergic neurons were injected in the lateral ventricles with 6-hydroxydopamine, a toxic that inhibits the mitochondrial chain of dopaminergic neurons and blockades the axonal transport. Dopaminergic terminals closest to the lateral ventricle showed an axonal fragmentation and an accumulation of damaged mitochondria in 2-9 μ saccular structures (spheroids). Damaged mitochondria accumulated in spheroids initiated (showing high Pink1, parkin, ubiquitin, p-S65-Ubi, AMBRA1, and BCL2L13 immunoreactivity and developing autophagosomes) but did not complete (mitochondria were not polyubiquitinated, autophagosomes had no STX17, and no lysosomes were found in spheroids) the mitophagy process. Then, spheroids were penetrated by astrocytic processes and DAergic mitochondria were transferred to astrocytes where they were polyubiquitinated (UbiK63+) and linked to mature autophagosomes (STX17+) which became autophagolysosomes (Lamp1/Lamp2 which co-localized with LC3). Present data provide evidence that the mitophagy of degenerating dopaminergic terminals starts in the dopaminergic spheroids and finishes in the surrounding astrocytes (spheroid-mediated transmitophagy). The neuron-astrocyte transmitophagy could be critical for preventing the release of damaged mitochondria to the extracellular medium and the neuro-inflammatory activity which characterizes Parkinson's disease.
Collapse
Affiliation(s)
- Ingrid Morales
- Laboratory of Neurobiology and Experimental Neurology, Department of Basic Medical Sciences, La Laguna University, La Laguna, Tenerife, Spain.,Center for Networked Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Alberto Sanchez
- Laboratory of Neurobiology and Experimental Neurology, Department of Basic Medical Sciences, La Laguna University, La Laguna, Tenerife, Spain
| | - Ricardo Puertas-Avendaño
- Laboratory of Neurobiology and Experimental Neurology, Department of Basic Medical Sciences, La Laguna University, La Laguna, Tenerife, Spain
| | | | - Adrian Perez-Barreto
- Laboratory of Neurobiology and Experimental Neurology, Department of Basic Medical Sciences, La Laguna University, La Laguna, Tenerife, Spain
| | - Manuel Rodriguez
- Laboratory of Neurobiology and Experimental Neurology, Department of Basic Medical Sciences, La Laguna University, La Laguna, Tenerife, Spain.,Center for Networked Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| |
Collapse
|
41
|
Validation of an in vivo electrochemical immunosensing platform for simultaneous detection of multiple cytokines in Parkinson's disease mice model. Bioelectrochemistry 2020; 134:107532. [PMID: 32305864 DOI: 10.1016/j.bioelechem.2020.107532] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 01/05/2023]
Abstract
Parkinson's Disease (PD) is a neurodegenerative chronic disorder which destroys brain tissue and result in impaired movement. Early diagnosis of PD before the appearance of clinical symptom is vital for effective treatment. High levels of proinflammatory cytokines found in PD patient's brains, as natural inflammation response product, are potential biomarkers for PD detection in the early stage. Herein, we developed an in vivo electrochemical immunosensing device based on glassy carbon rod to simultaneously detect three proinflammatory cytokines (IL-1β, IL-6 and TNF-α). The levels of IL-1β, IL-6 and TNF-α secreted by N2a cells significantly increased within 24 h after lipopolysaccharide (LPS) stimulation. Under in vivo conditions, the concentrations of IL-1β, IL-6 and TNF-α in PD model group achieved by injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intraperitoneally, were significantly higher than those in the control mouse group. The concentrations of three cytokines in vivo/vitro detected by this immunosensing device was comparable to that obtained by ELISA. Furthermore, this deployable immunosensing device was proved to be highly sensitive with the limits of detection (LODs) of 5 pg mL-1 for each cytokine, specific and reliable, suggesting its potential to be a universal immunosensing platform for early identification and diagnosis of PD in vivo in the future.
Collapse
|
42
|
Fu P, Gao M, Yung KKL. Association of Intestinal Disorders with Parkinson's Disease and Alzheimer's Disease: A Systematic Review and Meta-Analysis. ACS Chem Neurosci 2020; 11:395-405. [PMID: 31876406 DOI: 10.1021/acschemneuro.9b00607] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) and Alzheimer's disease (AD) are the most common neurodegenerative disorders, with an overall global incidence of 40 million. Many studies have revealed the association of intestinal disorders and bacterial infections with PD, but few studies have found such a relationship with AD. In this meta-analysis, related articles published up to September 2018 were searched in PubMed. Of the 2121 related articles screened initially, 56 were found to be eligible. Data on the risks of PD and AD due to five intestinal disorders and infection with Helicobacter pylori, as a representative intestinal microbe, were obtained, and a fixed- or random-effects model was used to pool the odds ratios (ORs) with 95% confidence interval (CIs) from individual studies. The combined OR for all types of intestinal disorders with an increased risk of PD was 3.36 (95% CI: 2.70-4.17). The ORs for each category were as follows: constipation, 4.05 (95% CI, 3.24-5.06); inflammatory bowel disease (IBD), 1.16 (95% CI, 0.89-1.52); irritable bowel syndrome (IBS), 1.75 (95% CI, 0.55-5.56); small intestinal bacterial overgrowth, 5.15 (95% CI, 3.33-7.96); and diarrhea, 1.27 (95% CI, 0.28-5.75). The combined OR of all types of intestinal disorders with an increased risk of AD was 1.52 (95% CI, 1.09-2.13). The ORs for IBS and IBD were 1.42 (95% CI, 1.02-1.99) and 2.40 (95% CI, 1.00-5.76), respectively. The risk estimates of H. pylori infection in PD and AD patients were as follows: OR, 1.65 (95% CI, 1.43-1.91) and OR, 1.40 (95% CI, 1.12-1.76), respectively. These findings suggest that PD and AD are significantly associated with intestinal disorders. The negative roles of H. pylori in the development of PD or AD should be evaluated to shed new light on the diagnosis and treatment of PD and AD. National governments should periodically inspect the intestinal condition of residents and extend health plans to improve intestinal health to prevent potential neurological disorders.
Collapse
Affiliation(s)
- Pengfei Fu
- Department of Biology, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong SAR, China
| | - Meng Gao
- Department of Geography, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong SAR, China
| | - Ken Kin Lam Yung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong SAR, China
| |
Collapse
|
43
|
Zahra W, Rai SN, Birla H, Singh SS, Rathore AS, Dilnashin H, Singh R, Keswani C, Singh RK, Singh SP. Neuroprotection of Rotenone-Induced Parkinsonism by Ursolic Acid in PD Mouse Model. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2020; 19:527-540. [PMID: 32787765 DOI: 10.2174/1871527319666200812224457] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Parkinson's Disease (PD) is characterized by both motor and non-motor symptoms. The presynaptic neuronal protein, α-Synuclein, plays a pivotal role in PD pathogenesis and is associated with both genetic and sporadic origin of the disease. Ursolic Acid (UA) is a well-known bioactive compound found in various medicinal plants, widely studied for its anti-inflammatory and antioxidant activities. OBJECTIVE In this research article, the neuroprotective potential of UA has been further explored in the Rotenone-induced mouse model of PD. METHODS To investigate our hypothesis, we have divided mice into 4 different groups, control, drug only control, Rotenone-intoxicated group, and Rotenone-intoxicated mice treated with UA. After the completion of dosing, behavioral parameters were estimated. Then mice from each group were sacrificed and the brains were isolated. Further, the biochemical tests were assayed to check the balance between the oxidative stress and endogenous anti-oxidants; and TH (Tyrosine Hydroxylase), α-Synuclein, Akt (Serine-threonine protein kinase), ERK (Extracellular signal-regulated kinase) and inflammatory parameters like Nuclear Factor-κB (NF-κB) and Tumor Necrosis Factor- α (TNF-α) were assessed using Immunohistochemistry (IHC). Western blotting was also done to check the expressions of TH and α-Synuclein. Moreover, the expression levels of PD related genes like α-Synuclein, β-Synuclein, Interleukin-1β (IL-1β), and Interleukin-10 (IL-10) were assessed by using Real-time PCR. RESULTS The results obtained in our study suggested that UA significantly reduced the overexpression of α-Synuclein and regulated the phosphorylation of survival-related kinases (Akt and ERK) apart from alleviating the behavioral abnormalities and protecting the dopaminergic neurons from oxidative stress and neuroinflammation. CONCLUSION Thus, our study shows the neuroprotective potential of UA, which can further be explored for possible clinical intervention.
Collapse
Affiliation(s)
- Walia Zahra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Sachchida Nand Rai
- Centre of Biotechnology, University of Allahabad, Prayagraj-211002, India
| | - Hareram Birla
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Saumitra Sen Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Aaina Singh Rathore
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Hagera Dilnashin
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Richa Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Chetan Keswani
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Rakesh K Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India
| |
Collapse
|
44
|
Zhu YF, Wang WP, Zheng XF, Chen Z, Chen T, Huang ZY, Jia LJ, Lei WL. Characteristic response of striatal astrocytes to dopamine depletion. Neural Regen Res 2020; 15:724-730. [PMID: 31638097 PMCID: PMC6975155 DOI: 10.4103/1673-5374.266917] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Astrocytes and astrocyte-related proteins play important roles in maintaining normal brain function, and also regulate pathological processes in brain diseases and injury. However, the role of astrocytes in the dopamine-depleted striatum remains unclear. A rat model of Parkinson’s disease was therefore established by injecting 10 μL 6-hydroxydopamine (2.5 μg/μL) into the right medial forebrain bundle. Immunohistochemical staining was used to detect the immunoreactivity of glial fibrillary acidic protein (GFAP), calcium-binding protein B (S100B), and signal transducer and activator of transcription 3 (STAT3) in the striatum, and to investigate the co-expression of GFAP with S100B and STAT3. Western blot assay was used to measure the protein expression of GFAP, S100B, and STAT3 in the striatum. Results demonstrated that striatal GFAP-immunoreactive cells had an astrocytic appearance under normal conditions, but that dopamine depletion induced a reactive phenotype with obvious morphological changes. The normal striatum also contained S100B and STAT3 expression. S100B-immunoreactive cells were uniform in the striatum, with round bodies and sparse, thin processes. STAT3-immunoreactive cells presented round cell bodies with sparse processes, or were darkly stained with a large cell body. Dopamine deprivation induced by 6-hydroxydopamine significantly enhanced the immunohistochemical positive reaction of S100B and STAT3. Normal striatal astrocytes expressed both S100B and STAT3. Striatal dopamine deprivation increased the number of GFAP/S100B and GFAP/STAT3 double-labeled cells, and increased the protein levels of GFAP, S100B, and STAT3. The present results suggest that morphological changes in astrocytes and changes in expression levels of astrocyte-related proteins are involved in the pathological process of striatal dopamine depletion. The study was approved by Animal Care and Use Committee of Sun Yat-sen University, China (Zhongshan Medical Ethics 2014 No. 23) on September 22, 2014.
Collapse
Affiliation(s)
- Yao-Feng Zhu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province; Institute of Medicine, College of Medicine, Jishou University, Jishou, Hunan Province, China
| | - Wei-Ping Wang
- Periodical Center, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xue-Feng Zheng
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhi Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Tao Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zi-Yun Huang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lin-Ju Jia
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Wan-Long Lei
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
45
|
Park T, Chen H, Kim HY. GPR110 (ADGRF1) mediates anti-inflammatory effects of N-docosahexaenoylethanolamine. J Neuroinflammation 2019; 16:225. [PMID: 31730008 PMCID: PMC6858791 DOI: 10.1186/s12974-019-1621-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 10/22/2019] [Indexed: 02/08/2023] Open
Abstract
Background Neuroinflammation is a widely accepted underlying condition for various pathological processes in the brain. In a recent study, synaptamide, an endogenous metabolite derived from docosahexaenoic acid (DHA, 22:6n-3), was identified as a specific ligand to orphan adhesion G-protein-coupled receptor 110 (GPR110, ADGRF1). Synaptamide has been shown to suppress lipopolysaccharide (LPS)-induced neuroinflammation in mice, but involvement of GPR110 in this process has not been established. In this study, we investigated the possible immune regulatory role of GPR110 in mediating the anti-neuroinflammatory effects of synaptamide under a systemic inflammatory condition. Methods For in vitro studies, we assessed the role of GPR110 in synaptamide effects on LPS-induced inflammatory responses in adult primary mouse microglia, immortalized murine microglial cells (BV2), primary neutrophil, and peritoneal macrophage by using quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA) as well as neutrophil migration and ROS production assays. To evaluate in vivo effects, wild-type (WT) and GPR110 knock-out (KO) mice were injected with LPS intraperitoneally (i.p.) or TNF intravenously (i.v.) followed by synaptamide (i.p.), and expression of proinflammatory mediators was measured by qPCR, ELISA, and western blot analysis. Activated microglia in the brain and NF-kB activation in cells were examined microscopically after immunostaining for Iba-1 and RelA, respectively. Results Intraperitoneal (i.p.) administration of LPS increased TNF and IL-1β in the blood and induced pro-inflammatory cytokine expression in the brain. Subsequent i.p. injection of the GPR110 ligand synaptamide significantly reduced LPS-induced inflammatory responses in wild-type (WT) but not in GPR110 knock-out (KO) mice. In cultured microglia, synaptamide increased cAMP and inhibited LPS-induced proinflammatory cytokine expression by inhibiting the translocation of NF-κB subunit RelA into the nucleus. These effects were abolished by blocking synaptamide binding to GPR110 using an N-terminal targeting antibody. GPR110 expression was found to be high in neutrophils and macrophages where synaptamide also caused a GPR110-dependent increase in cAMP and inhibition of LPS-induced pro-inflammatory mediator expression. Intravenous injection of TNF, a pro-inflammatory cytokine that increases in the circulation after LPS treatment, elicited inflammatory responses in the brain which were dampened by the subsequent injection (i.p.) of synaptamide in a GPR110-dependent manner. Conclusion Our study demonstrates the immune-regulatory function of GPR110 in both brain and periphery, collectively contributing to the anti-neuroinflammatory effects of synaptamide under a systemic inflammatory condition. We suggest GPR110 activation as a novel therapeutic strategy to ameliorate inflammation in the brain as well as periphery.
Collapse
Affiliation(s)
- Taeyeop Park
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, 5625 Fishers Lane, Rm. 3N-07, Rockville, MD, 20852, USA
| | - Huazhen Chen
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, 5625 Fishers Lane, Rm. 3N-07, Rockville, MD, 20852, USA
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, 5625 Fishers Lane, Rm. 3N-07, Rockville, MD, 20852, USA.
| |
Collapse
|
46
|
Methyl jasmonate abrogates rotenone-induced parkinsonian-like symptoms through inhibition of oxidative stress, release of pro-inflammatory cytokines, and down-regulation of immnopositive cells of NF-κB and α-synuclein expressions in mice. Neurotoxicology 2019; 74:172-183. [DOI: 10.1016/j.neuro.2019.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 06/12/2019] [Accepted: 07/15/2019] [Indexed: 01/07/2023]
|
47
|
Msibi ZNP, Mabandla MV. Oleanolic Acid Mitigates 6-Hydroxydopamine Neurotoxicity by Attenuating Intracellular ROS in PC12 Cells and Striatal Microglial Activation in Rat Brains. Front Physiol 2019; 10:1059. [PMID: 31496954 PMCID: PMC6712087 DOI: 10.3389/fphys.2019.01059] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 08/02/2019] [Indexed: 12/30/2022] Open
Abstract
Oleanolic acid (OA), a biologically active pentacyclic triterpenoid compound, has been implicated in a number of clinical benefits including antioxidant, and anti-inflammatory properties. OA has been previously shown to ameliorate the toxic effects of 6-hydroxydopamine (6-OHDA), however, the mechanism by which this effect is exhibited is not clearly understood. In the present study, we investigated the role of OA in attenuation of microglial activation in 6-OHDA induced Parkinsonian rat model. We also explored the ability of OA to attenuate 6-OHDA-induced intracellular reactive oxygen species (ROS), and thus prevent cell death in PC12 cells. We accessed the utility of immunohistochemistry to assess striatal microglial activation, where shape descriptors such as area, perimeter, Feret's diameter, aspect ratio and solidity were determined using the Fiji ImageJ software. Intracellular ROS and cell viability were assessed in PC12 cells using the OxiSelectTM Intracellular ROS Assay Kit and MTT assay, respectively. We found that microglial activation was decreased in rats pre-treated with OA prior to 6-OHDA insult as well as in rats treated with OA 1 day post 6-OHDA exposure when compared to untreated rats, as determined by shape descriptors. This finding was in correlation with significantly improved motor symptoms and increased striatal dopamine in treated rats as compared to non-treated rats. Flow cytometry assessment of PC12 cells revealed a decreased amount of intracellular ROS in cells pre-treated with OA 6 h prior to 6-OHDA exposure and cells treated with OA 1 h post 6-OHDA exposure, suggesting that OA provides neuroprotection in PC12 cells by removing intracellular ROS, thereby reducing oxidative stress. Our finding suggest that OA exhibits its neuroprotective effect by attenuating striatal microglial activation, which results in neuroinflammation that is implicated in Parkinson's disease pathology. Further studies detailing the mechanism by which OA interacts with microglia may be useful in understanding the role of OA in attenuating neuroinflammation.
Collapse
Affiliation(s)
- Zama N P Msibi
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Musa V Mabandla
- Laboratory Medicine and Medical Sciences, Nelson R Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
48
|
Kim KH, Kim M, Lee J, Jeon HN, Kim SH, Bae H. Comparison of the Protective Effects of Bee Venom Extracts with Varying PLA 2 Compositions in a Mouse Model of Parkinson's Disease. Toxins (Basel) 2019; 11:toxins11060358. [PMID: 31248167 PMCID: PMC6628630 DOI: 10.3390/toxins11060358] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/13/2019] [Accepted: 06/15/2019] [Indexed: 12/15/2022] Open
Abstract
Bee venom contains a number of pharmacologically active components, including enzymes and polypeptides such as phospholipase A2 (PLA2) and melittin, which have been shown to exhibit therapeutic benefits, mainly via attenuation of inflammation, neurotoxicity, and nociception. The individual components of bee venom may manifest distinct biological actions and therapeutic potential. In this study, the potential mechanisms of action of PLA2 and melittin, among different compounds purified from honey bee venom, were evaluated against Parkinson’s disease (PD). Notably, bee venom PLA2 (bvPLA2), but not melittin, exhibited neuroprotective activity against PD in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. MPTP-induced behavioral deficits were also abolished after bvPLA2 treatment, depending on the PLA2 content. Further, bvPLA2 administration activated regulatory T cells (Tregs) while inhibiting inflammatory T helper (Th) 1 and Th17 cells in the MPTP mouse model of PD. These results indicate that bvPLA2, but not melittin, protected against MPTP and alleviated inflammation in PD. Thus, bvPLA2 is a promising and effective therapeutic agent in Parkinson’s disease.
Collapse
Affiliation(s)
- Kyung Hwa Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.
| | - Minhwan Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.
| | - Jaehwan Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.
| | - Hat Nim Jeon
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.
| | - Se Hyun Kim
- Inist ST Co. Ltd., 159 Sagimakgol-ro, Jungwon-gu, Seongnam-si, Gyeonggi-do 13202, Korea.
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.
| |
Collapse
|
49
|
Cohen J, Torres C. Astrocyte senescence: Evidence and significance. Aging Cell 2019; 18:e12937. [PMID: 30815970 PMCID: PMC6516680 DOI: 10.1111/acel.12937] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/14/2019] [Accepted: 02/04/2019] [Indexed: 12/12/2022] Open
Abstract
Astrocytes participate in numerous aspects of central nervous system (CNS) physiology ranging from ion balance to metabolism, and disruption of their physiological roles can therefore be a contributor to CNS dysfunction and pathology. Cellular senescence, one of the mechanisms of aging, has been proposed as a central component of the age dependency of neurodegenerative disorders. Cumulative evidence supports an integral role of astrocytes in the initiation and progression of neurodegenerative disease and cognitive decline with aging. The loss of astrocyte function or the gain of neuroinflammatory function as a result of cellular senescence could have profound implications for the aging brain and neurodegenerative disorders, and we propose the term “astrosenescence” to describe this phenotype. This review summarizes the current evidence pertaining to astrocyte senescence from early evidence, in vitro characterization and relationship to age‐related neurodegenerative disease. We discuss the significance of targeting senescent astrocytes as a novel approach toward therapies for age‐associated neurodegenerative disease.
Collapse
Affiliation(s)
- Justin Cohen
- Department of Pathology and Laboratory Medicine Drexel University College of Medicine Philadelphia Pennsylvania
| | - Claudio Torres
- Department of Pathology and Laboratory Medicine Drexel University College of Medicine Philadelphia Pennsylvania
| |
Collapse
|
50
|
Fu P, Guo X, Cheung FMH, Yung KKL. The association between PM 2.5 exposure and neurological disorders: A systematic review and meta-analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 655:1240-1248. [PMID: 30577116 DOI: 10.1016/j.scitotenv.2018.11.218] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 05/11/2023]
Abstract
BACKGROUND Recent systematic review and meta-analyses have tried to identify an association between PM2.5 exposure and stroke, but few could find a conclusive and comprehensive evidence. Moreover, the associations between PM2.5, neurodegenerative diseases and neurodevelopmental disorders have never been reviewed. We aimed to assess the effects of PM2.5 exposure on stroke, dementia, Alzheimer's disease, autism spectrum disorder (ASD), Parkinson's disease, and mild cognitive impairment (MCI). METHODS We searched PubMed and CNKI databases for articles published until June 2018. Studies were eligible for analysis if they were human studies and provided risk estimates with 95% CI. We screened 1645 articles and identified 80 eligible studies covering 26 countries across all continents except Antarctica. Risks of incidence and mortality were extracted and stratified by types of neurological disorders, PM2.5 concentration and duration of PM2.5 exposure. RESULTS We found significant association between PM2.5 exposure and stroke, dementia, Alzheimer's disease, ASD, Parkinson's disease. The risks of ischemic and hemorrhagic stroke were higher than that of stroke in general, and that hemorrhagic stroke had by far the highest mortality. The risk of stroke for heavily polluted countries was significantly higher than that of lightly polluted countries. Short- and long-term PM2.5 exposure was associated with increased risks of stroke (short-term odds ratio 1.01 [per 10 μg/m3 increase in PM2.5 concentrations], 95% CI 1.01-1.02; long-term 1.14, 95% CI 1.08-1.21) and mortality (short-term 1.02, 95% CI 1.01-1.04; long-term 1.15, 95% CI 1.07-1.24) of stroke. Long-term PM2.5 exposure was associated with increased risks of dementia (1.16, 95% CI 1.07-1.26), Alzheimer's disease (3.26, 95% 0.84-12.74), ASD (1.68, 95% CI 1.20-2.34), and Parkinson's disease (1.34, 95% CI 1.04-1.73). CONCLUSIONS There is a strong association between PM2.5 exposure and neurological disorders. National governments should exert greater efforts to improve air quality given its health implications.
Collapse
Affiliation(s)
- Pengfei Fu
- Department of Biology, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Xinbiao Guo
- Department of Occupational & Environmental Health Sciences, Peking University School of Public Health, Beijing, China
| | | | - Ken Kin Lam Yung
- Department of Biology, Hong Kong Baptist University, Hong Kong, SAR, China.
| |
Collapse
|