1
|
Xie L, Zhuang Z, Guo B, Huang Y, Shi X, Huang Z, Xu Z, Chen Y, Cao Y, Zheng Y, Wu R, Ma S. Ketamine induced gut microbiota dysbiosis and barrier and hippocampal dysfunction in rats. iScience 2024; 27:111089. [PMID: 39493883 PMCID: PMC11530865 DOI: 10.1016/j.isci.2024.111089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/02/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
The microbiota-gut-brain axis (MGBA) plays a pivotal role in drug addiction. However, the pathophysiological mechanism of MGBA in ketamine addiction remains elusive. The present study investigated the ketamine-induced gut microbiota disorders, intestinal barrier dysfunction, and the alterations in brain function, using a conditioned place preference (CPP) model of ketamine addiction in rats. Compared with the control group, ketamine induced decreased amplitude of low-frequency fluctuation (ALFF) values in the hippocampus, and pyknotic nuclei and concentrated cytoplasm in hippocampal neurons, as well as alterations in gut microbiota composition, shortened ileum villi, and thinner colonic mucosa. We also found that the abundance of gut microbiota exhibited correlations with CPP score, hippocampal ALFF value, length of ileum villi, and thickness of colonic mucosa. Our findings provide evidence for abnormal alterations in the MGBA of ketamine-addicted rats, which improves our understating of the mechanism of ketamine addiction and the potential for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Lei Xie
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Zelin Zhuang
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Baowen Guo
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yuehua Huang
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Xiaoyan Shi
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Zikai Huang
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Ziquan Xu
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yanbin Chen
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yuyin Cao
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yanmin Zheng
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Renhua Wu
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, China
| | - Shuhua Ma
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
2
|
Kim B, Yang M, Lee J, Kim JS, Hyun SH, Moon C. Upregulation of γ-synuclein in the prefrontal cortex and hippocampus following dopamine depletion: A study using the striatal 6-hydroxydopamine hemiparkinsonian rat model. Neurosci Lett 2024; 839:137936. [PMID: 39151573 DOI: 10.1016/j.neulet.2024.137936] [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: 04/26/2024] [Revised: 07/16/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Synucleins, including α-synuclein (α-syn), β-syn, and γ-syn, have been implicated in various synucleinopathies, notably Parkinson's disease (PD), which has generated increased interest in understanding their roles. Although α-syn and β-syn have contrasting neuropathological consequences, the precise role of γ-syn remains unclear. This study validated non-motor symptoms, specifically anxiety-like behavior, along with the degradation of dopaminergic (DAergic) neurons in the nigrostriatal system and DAergic neurites in the prefrontal cortex and hippocampus of rats infused with striatal 6-hydroxydopamine (6-OHDA). Our study further investigated the alterations in γ-syn expression levels in the prefrontal cortices and hippocampi of these 6-OHDA-treated rats, aiming to establish foundational insights into the neuropathophysiology of DA depletion, a central feature of PD. Our findings revealed a significant increase in the expression of γ-syn mRNA and protein in these brain regions, in contrast to unaltered α- and β-syn expression levels. This suggests a distinct role of γ-syn within the neurobiological milieu under conditions of DA deficiency. Overall, our data shed light on the neurobiological changes observed in the hemiparkinsonian rat model induced with 6-OHDA, underscoring the potential significance of γ-syn in PD pathology.
Collapse
Affiliation(s)
- Bohye Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Miyoung Yang
- Department of Anatomy, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Jeongmin Lee
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
3
|
Mishra J, Walecha V, Sophronea T, Singh A, Agrawal S, Luthra PM. BBPT attenuated 6-OHDA-induced toxicity by modulating oxidative stress, apoptotic, and inflammatory proteins in primary neurons and rat models of Parkinson's disease. Neurotoxicology 2024; 105:67-81. [PMID: 39216605 DOI: 10.1016/j.neuro.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Parkinson's disease (PD) results from the degeneration of dopaminergic neurons in substantia nigra pars compacta (SNpc). Adenosine A2AR acting through the striato-pallidal pathway has emerged as a non-dopaminergic target in the therapy of PD. In the present work, the anti-parkinsonian potential of (4E)-4-(4-bromobenzylideneamino)-3-phenyl-2,3-dihydro-2-thioxo- thiazole-5-carbonitrile (BBPT) was explored. BBPT exhibited significant antioxidant activity in situ. In the MTT assay, the BBPT treatment showed insignificant toxicity to the primary midbrain neuronal (PMDN) cells. 6-OHDA induced PMDN cells, 3 h post-treated with BBPT showed 80-85 % survival of the cells and restoration of dopamine and TNF-α levels. The acute and sub-acute toxicity test for BBPT was performed with Sprague Dawley (SD) rats. In toxicity assay, any significant physical, hematological, or biochemical changes in the rats were not observed. To evaluate the effect of BBPT in vivo, a 6-OHDA-induced unilaterally lesioned SD rat model of PD was established. We observed that the BBPT treatment improved the behavioral symptoms in 6-OHDA-induced unilaterally lesioned rats. The proteins of 6-OHDA-induced BBPT-treated rats were isolated from the brain tissue to assess the antioxidant effect (GSH, catalase, SOD, lipid-peroxidation, nitrite), dopamine levels, and the restoration in the apoptosis and inflammation. Our results demonstrated that BBPT increased the anti-oxidant enzyme levels, restored the caspase-3/Bcl-2 levels to arrest apoptosis, and attenuated the TNF-α/IL-6 levels, thus restoring the neuronal damage in unilaterally lesioned 6-OHDA-induced SD rats. Precisely, the findings suggested that BBPT possessed significant anti-parkinsonian activity and has the potential to prevent dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Jyoti Mishra
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi 110007, India
| | - Vaishali Walecha
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi 110007, India
| | - Tuithung Sophronea
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi 110007, India
| | - Ankit Singh
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi 110007, India
| | - Saurabh Agrawal
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi 110007, India
| | - Pratibha Mehta Luthra
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi 110007, India.
| |
Collapse
|
4
|
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
|
5
|
Yang J, Lin W, Ma Y, Song H, Mu C, Wu Q, Han C, Zhang J, Liu X. Investigation of the causal association between Parkinson's disease and autoimmune disorders: a bidirectional Mendelian randomization study. Front Immunol 2024; 15:1370831. [PMID: 38774879 PMCID: PMC11106379 DOI: 10.3389/fimmu.2024.1370831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/24/2024] [Indexed: 05/24/2024] Open
Abstract
Background To date, an increasing number of epidemiological evidence has pointed to potential relationships between Parkinson's disease (PD) and various autoimmune diseases (AIDs), however, no definitive conclusions has been drawn about whether PD is causally related to AIDs risk. Methods By employing summary statistics from the latest and most extensive genome-wide association studies (GWAS), we performed a bidirectional two-sample Mendelian randomization (MR) analysis to investigate the causal associations between PD and a variety of 17 AIDs, encompassing multiple sclerosis, neuromyelitis optica spectrum disorder, myasthenia gravis, asthma, inflammatory bowel disease, Crohn's disease, ulcerative colitis, irritable bowel syndrome, celiac disease, primary biliary cirrhosis, primary sclerosing cholangitis, type 1 diabetes, ankylosing spondylitis, rheumatoid arthritis, systemic lupus erythematosus, psoriasis and vitiligo. Inverse-variance weighted (IVW) was adopted as the main statistical approach to obtain the causal estimates of PD on different AIDs, supplemented by a series of complementary analyses (weighted median, MR Egger regression, and MR-PRESSO) for further strengthening the robustness of results. Results Our MR findings suggested that genetically predicted higher liability to PD was causally associated with a decreased risk of irritable bowel syndrome (OR = 0.98; 95% CI: 0.96-0.99; P = 0.032). On the contrary, IVW analysis showed a potential positive correlation between genetically determined PD and the incidence of type 1 diabetes (OR = 1.10; 95%CI: 1.02-1.19; P = 0.010). Subsequent MR tests ended up in similar results, confirming our findings were reliable. Additionally, in the reverse MR analyses, we did not identify any evidence to support the causal relationship of genetic predisposition to AIDs with PD susceptibility. Conclusion In general, a bifunctional role that PD exerted on the risk of developing AIDs was detected in our studies, both protecting against irritable bowel syndrome occurrence and raising the incidence of type 1 diabetes. Future studies, including population-based observational studies and molecular experiments in vitro and in vivo, are warranted to validate the results of our MR analyses and refine the underlying pathological mechanisms involved in PD-AIDs associations.
Collapse
Affiliation(s)
- Junyi Yang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Weiran Lin
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the People's Republic of China, China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of the People's Republic of China, China Medical University, Shenyang, Liaoning, China
- Department of Laboratory Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Yumei Ma
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hui Song
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the People's Republic of China, China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of the People's Republic of China, China Medical University, Shenyang, Liaoning, China
| | - Changqing Mu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qian Wu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Han
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jian Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the People's Republic of China, China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Medical Cell Biology, Ministry of Education of the People's Republic of China, China Medical University, Shenyang, Liaoning, China
| | - Xu Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Cui D, Chen Y, Ye B, Guo W, Wang D, He J. Natural products for the treatment of neurodegenerative diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155101. [PMID: 37778246 DOI: 10.1016/j.phymed.2023.155101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 08/29/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Neurodegenerative diseases are among the most common diseases in older adults worldwide. Alzheimer's disease (AD) and Parkinson's disease (PD) are two of the most common neurodegenerative diseases, and are accompanied by cerebral cortical atrophy, neuronal loss, protein accumulation, and excessive accumulation of metal ions. Natural products exhibit outstanding performance in improving cerebral circulatory disorders, promoting cerebral haematoma absorption, repairing damaged nerve tissue, and improving damaged nerve function. In recent years, studies have shown that neuroinflammatory mechanisms and signalling pathways closely related to the occurrence and development of neurological diseases include microglial activation, nuclear factor-κB (NF-κB) pathway, mitogen activated protein kinases (MAPK) pathway, reactive oxygen pathway, nucleotide binding oligomerisation domain-like receptor protein3 (NLRP3) inflammasomes, toll-like receptor4 (TLR4) pathway, nuclear factor erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) pathway, phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, and intestinal flora. Therefore, this study considered the mechanism of neurological diseases as the starting point to review the mechanism of action of natural products in the prevention and treatment of AD and PD in recent years to provide a theoretical basis for clinical prevention and treatment. AIM Natural products are a promising source of novel lead structures that have long been used to treat various nervous system diseases. METHODOLOGY This review collected literature on neurological diseases and natural products from 2012 to 2022, which were mainly searched through databases such as ScienceDirect, Springer, PubMed, SciFinder, China National Knowledge Infrastructure (CNKI), Wanfang, Google Scholar, and Baidu Academic. The following keywords were searched: neurological disorders, natural products, signalling pathway, mechanism of action. RESULTS This review summarises the pathogenesis of degenerative neurological diseases, recent findings on natural products used in neurodegenerative diseases, and the molecular mechanisms underlying these effects.
Collapse
Affiliation(s)
- Donghan Cui
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University, West China Hospital, Chengdu 610041, China
| | - Yajuan Chen
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Bengui Ye
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China; Medical College of Tibet University, Lasa 850002, China
| | - Wenhao Guo
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University, West China Hospital, Chengdu 610041, China.
| | - Dongdong Wang
- Centre for Metabolism, Obesity, and Diabetes Research, Department of Medicine, McMaster University, HSC 4N71, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada.
| | - Jun He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China.
| |
Collapse
|
7
|
Vellingiri B, Balasubramani K, Iyer M, Raj N, Elangovan A, Song K, Yeo HC, Jayakumar N, Kinoshita M, Thangarasu R, Narayanasamy A, Dayem AA, Prajapati VK, Gopalakrishnan AV, Cho SG. Role of Telomeres and Telomerase in Parkinson's Disease-A New Theranostics? Adv Biol (Weinh) 2023; 7:e2300097. [PMID: 37590305 DOI: 10.1002/adbi.202300097] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/19/2023] [Indexed: 08/19/2023]
Abstract
Parkinson's disease (PD) is a complex condition that is significantly influenced by oxidative stress and inflammation. It is also suggested that telomere shortening (TS) is regulated by oxidative stress which leads to various diseases including age-related neurodegenerative diseases like PD. Thus, it is anticipated that PD would result in TS of peripheral blood mononuclear cells (PBMCs). Telomeres protect the ends of eukaryotic chromosomes preserving them against fusion and destruction. The TS is a normal process because DNA polymerase is unable to replicate the linear ends of the DNA due to end replication complications and telomerase activity in various cell types counteracts this process. PD is usually observed in the aged population and progresses over time therefore, disparities among telomere length in PBMCs of PD patients are recorded and it is still a question whether it has any useful role. Here, the likelihood of telomere attrition in PD and its implications concerning microglia activation, ageing, oxidative stress, and the significance of telomerase activators are addressed. Also, the possibility of telomeres and telomerase as a diagnostic and therapeutic biomarker in PD is discussed.
Collapse
Affiliation(s)
- Balachandar Vellingiri
- Stem Cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Kiruthika Balasubramani
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Mahalaxmi Iyer
- Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore, Tamil Nadu, 641021, India
| | - Neethu Raj
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Ajay Elangovan
- Stem Cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Kwonwoo Song
- Department of Stem Cell and Regenerative Biotechnology, Molecular and Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, 05029, Republic of Korea
| | - Han-Cheol Yeo
- Department of Stem Cell and Regenerative Biotechnology, Molecular and Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, 05029, Republic of Korea
| | - Namitha Jayakumar
- Department of Biotechnology, Sri Ramakrishna College of Arts and Science, Coimbatore, Tamil Nadu, 641006, India
| | - Masako Kinoshita
- Department of Neurology, National Hospital Organization Utano National Hospital, Ondoyama-Cho, Narutaki, Ukyo-Ku, Kyoto, 616-8255, Japan
| | - Ravimanickam Thangarasu
- Department of Zoology, School of Science, Tamil Nadu Open University, Saidapet, Chennai, 600015, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular and Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, 05029, Republic of Korea
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular and Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, 05029, Republic of Korea
| |
Collapse
|
8
|
Singh J, Habean ML, Panicker N. Inflammasome assembly in neurodegenerative diseases. Trends Neurosci 2023; 46:814-831. [PMID: 37633753 PMCID: PMC10530301 DOI: 10.1016/j.tins.2023.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/26/2023] [Accepted: 07/31/2023] [Indexed: 08/28/2023]
Abstract
Neurodegenerative disorders are characterized by the progressive dysfunction and death of selectively vulnerable neuronal populations, often associated with the accumulation of aggregated host proteins. Sustained brain inflammation and hyperactivation of inflammasome complexes have been increasingly demonstrated to contribute to neurodegenerative disease progression. Here, we review molecular mechanisms leading to inflammasome assembly in neurodegeneration. We focus primarily on four degenerative brain disorders in which inflammasome hyperactivation has been well documented: Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and the spectrum of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). We discuss shared and divergent principles of inflammasome assembly across these disorders, and underscore the differences between neurodegeneration-associated inflammasome activation pathways and their peripheral-immune counterparts. We examine how aberrant assembly of inflammasome complexes may amplify pathology in neurodegeneration, including misfolded protein aggregation, and highlight prospects for neurotherapeutic interventions based on targeting inflammasome pathways.
Collapse
Affiliation(s)
- Jagjit Singh
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Maria L Habean
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Biomedical Scientist Training Program (Department of Neurosciences), Case Western Reserve University, Cleveland, OH, USA
| | - Nikhil Panicker
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44106, USA; Kent State University, Neurosciences, School of Biomedical Sciences, Cleveland, OH, USA.
| |
Collapse
|
9
|
Thomas R, Connolly KJ, Brekk OR, Hinrich AJ, Hastings ML, Isacson O, Hallett PJ. Viral-like TLR3 induction of cytokine networks and α-synuclein are reduced by complement C3 blockade in mouse brain. Sci Rep 2023; 13:15164. [PMID: 37704739 PMCID: PMC10499893 DOI: 10.1038/s41598-023-41240-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023] Open
Abstract
Inflammatory processes and mechanisms are of central importance in neurodegenerative diseases. In the brain, α-synucleinopathies such as Parkinson's disease (PD) and Lewy body dementia (LBD) show immune cytokine network activation and increased toll like receptor 3 (TLR3) levels for viral double-stranded RNA (dsRNA). Brain inflammatory reactions caused by TLR3 activation are also relevant to understand pathogenic cascades by viral SARS-CoV-2 infection causing post- COVID-19 brain-related syndromes. In the current study, following regional brain TLR3 activation induced by dsRNA in mice, an acute complement C3 response was seen at 2 days. A C3 splice-switching antisense oligonucleotide (ASO) that promotes the splicing of a non-productive C3 mRNA, prevented downstream cytokines, such as IL-6, and α-synuclein changes. This report is the first demonstration that α-synuclein increases occur downstream of complement C3 activation. Relevant to brain dysfunction, post-COVID-19 syndromes and pathological changes leading to PD and LBD, viral dsRNA TLR3 activation in the presence of C3 complement blockade further revealed significant interactions between complement systems, inflammatory cytokine networks and α-synuclein changes.
Collapse
Affiliation(s)
- Ria Thomas
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Kyle J Connolly
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Oeystein R Brekk
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Anthony J Hinrich
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Ole Isacson
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA.
| | - Penelope J Hallett
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
10
|
Yu Z, Chen W, Zhang L, Chen Y, Chen W, Meng S, Lu L, Han Y, Shi J. Gut-derived bacterial LPS attenuates incubation of methamphetamine craving via modulating microglia. Brain Behav Immun 2023; 111:101-115. [PMID: 37004759 DOI: 10.1016/j.bbi.2023.03.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/16/2023] [Accepted: 03/28/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND The microbiota-gut-brain axis plays a critical role in the pathophysiology of neuropsychiatric disorders, and the compositions of gut microbiota are altered by addictive drugs. However, the role of gut microbiota in the incubation of methamphetamine (METH) craving remains poorly understood. METHODS 16S rRNA gene sequencing was performed to assess the richness and diversity of gut microbiota in METH self-administration model. Hematoxylin and eosin staining was performed to evaluate the integrity of intestinal barrier. Immunofluorescence and three-dimensional reconstruction were performed to assess the morphologic changes of microglia. Serum levels of lipopolysaccharide (LPS) were determined using the rat enzyme-linked immunosorbent assay kits. Quantitative real-time PCR was performed to assess transcript levels of dopamine receptor, glutamate ionotropic AMPA receptor 3 and brain-derived neurotrophic factor. RESULTS METH self-administration induced gut microbiota dysbiosis, intestinal barrier damage and microglia activation in the nucleus accumbens core (NAcc), which was partially recovered after prolonged withdrawal. Microbiota depletion via antibiotic treatment increased LPS levels and induced a marked change in the microglial morphology in the NAcc, as indicated by the decreases in the lengths and numbers of microglial branches. Depleting the gut microbiota also prevented the incubation of METH craving and increased the population of Klebsiella oxytoca. Furthermore, Klebsiella oxytoca treatment or exogenous administration of the gram-negative bacterial cell wall component LPS increased serum and central LPS levels, induced microglial morphological changes and reduced the dopamine receptor transcription in the NAcc. Both treatments and NAcc microinjections of gut-derived bacterial LPS significantly decreased METH craving after prolonged withdrawal. CONCLUSIONS These data suggest that LPS from gut gram-negative bacteria may enter circulating blood, activate microglia in the brain and consequently decrease METH craving after withdrawal, which may have important implications for novel strategies to prevent METH addiction and relapse.
Collapse
Affiliation(s)
- Zhoulong Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Wenjun Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Libo Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yun Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Wenxi Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China.
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking University Shenzhen Hospital, Shenzhen 518036, China; The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, Beijing 100191, China; The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| |
Collapse
|
11
|
Isik S, Yeman Kiyak B, Akbayir R, Seyhali R, Arpaci T. Microglia Mediated Neuroinflammation in Parkinson’s Disease. Cells 2023; 12:cells12071012. [PMID: 37048085 PMCID: PMC10093562 DOI: 10.3390/cells12071012] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Parkinson’s Disease (PD) is the second most common neurodegenerative disorder seen, especially in the elderly. Tremor, shaking, movement problems, and difficulty with balance and coordination are among the hallmarks, and dopaminergic neuronal loss in substantia nigra pars compacta of the brain and aggregation of intracellular protein α-synuclein are the pathological characterizations. Neuroinflammation has emerged as an involving mechanism at the initiation and development of PD. It is a complex network of interactions comprising immune and non-immune cells in addition to mediators of the immune response. Microglia, the resident macrophages in the CNS, take on the leading role in regulating neuroinflammation and maintaining homeostasis. Under normal physiological conditions, they exist as “homeostatic” but upon pathological stimuli, they switch to the “reactive state”. Pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes are used to classify microglial activity with each phenotype having its own markers and released mediators. When M1 microglia are persistent, they will contribute to various inflammatory diseases, including neurodegenerative diseases, such as PD. In this review, we focus on the role of microglia mediated neuroinflammation in PD and also signaling pathways, receptors, and mediators involved in the process, presenting the studies that associate microglia-mediated inflammation with PD. A better understanding of this complex network and interactions is important in seeking new therapies for PD and possibly other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sevim Isik
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Uskudar, Istanbul 34662, Turkey
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Correspondence: ; Tel.: +90-216-400-2222 (ext. 2462)
| | - Bercem Yeman Kiyak
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Medicine, Institute of Hamidiye Health Sciences, University of Health Sciences, Uskudar, Istanbul 34668, Turkey
| | - Rumeysa Akbayir
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Rama Seyhali
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Tahire Arpaci
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| |
Collapse
|
12
|
Basurco L, Abellanas MA, Ayerra L, Conde E, Vinueza-Gavilanes R, Luquin E, Vales A, Vilas A, Martin-Uriz PS, Tamayo I, Alonso MM, Hernaez M, Gonzalez-Aseguinolaza G, Clavero P, Mengual E, Arrasate M, Hervás-Stubbs S, Aymerich MS. Microglia and astrocyte activation is region-dependent in the α-synuclein mouse model of Parkinson's disease. Glia 2023; 71:571-587. [PMID: 36353934 PMCID: PMC10100513 DOI: 10.1002/glia.24295] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/16/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
Inflammation is a common feature in neurodegenerative diseases that contributes to neuronal loss. Previously, we demonstrated that the basal inflammatory tone differed between brain regions and, consequently, the reaction generated to a pro-inflammatory stimulus was different. In this study, we assessed the innate immune reaction in the midbrain and in the striatum using an experimental model of Parkinson's disease. An adeno-associated virus serotype 9 expressing the α-synuclein and mCherry genes or the mCherry gene was administered into the substantia nigra. Myeloid cells (CD11b+ ) and astrocytes (ACSA2+ ) were purified from the midbrain and striatum for bulk RNA sequencing. In the parkinsonian midbrain, CD11b+ cells presented a unique anti-inflammatory transcriptomic profile that differed from degenerative microglia signatures described in experimental models for other neurodegenerative conditions. By contrast, striatal CD11b+ cells showed a pro-inflammatory state and were similar to disease-associated microglia. In the midbrain, a prominent increase of infiltrated monocytes/macrophages was observed and, together with microglia, participated actively in the phagocytosis of dopaminergic neuronal bodies. Although striatal microglia presented a phagocytic transcriptomic profile, morphology and cell density was preserved and no active phagocytosis was detected. Interestingly, astrocytes presented a pro-inflammatory fingerprint in the midbrain and a low number of differentially displayed transcripts in the striatum. During α-synuclein-dependent degeneration, microglia and astrocytes experience context-dependent activation states with a different contribution to the inflammatory reaction. Our results point towards the relevance of selecting appropriate cell targets to design neuroprotective strategies aimed to modulate the innate immune system during the active phase of dopaminergic degeneration.
Collapse
Affiliation(s)
- Leyre Basurco
- Departamentode Bioquímica y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain.,Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Miguel Angel Abellanas
- Departamentode Bioquímica y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain.,Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Leyre Ayerra
- Departamentode Bioquímica y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain.,Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Enrique Conde
- Programa de Inmunología, CIMA-Universidad de Navarra, Pamplona, Spain
| | | | - Esther Luquin
- Departamento de Patología, Anatomía y Fisiología, Facultad de Medicina, Universidad de Navarra, Pamplona, Spain
| | - Africa Vales
- Programa de Terapia Génica, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Amaya Vilas
- Programa de Oncohematología, CIMA-Universidad de Navarra, Pamplona, Spain
| | | | - Ibon Tamayo
- Programa de Biología Computacional, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Marta M Alonso
- Programa de Tumores Sólidos, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| | - Mikel Hernaez
- Programa de Biología Computacional, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Gloria Gonzalez-Aseguinolaza
- Programa de Terapia Génica, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| | - Pedro Clavero
- Servicio de Neurología, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Elisa Mengual
- Departamento de Patología, Anatomía y Fisiología, Facultad de Medicina, Universidad de Navarra, Pamplona, Spain
| | - Montserrat Arrasate
- Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| | - Sandra Hervás-Stubbs
- Programa de Inmunología, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| | - Maria S Aymerich
- Departamentode Bioquímica y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain.,Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain.,Programa de Tumores Sólidos, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| |
Collapse
|
13
|
Eyvari Brooshghalan S, Sabahi M, Ebadi SA, Sadeghian Z, Mohajjel Nayebi A, Haddadi R. Silibinin chronic treatment in a rat model of Parkinson disease: A comprehensive in-vivo evaluation and in silico molecular modeling. Eur J Pharmacol 2023; 941:175517. [PMID: 36669615 DOI: 10.1016/j.ejphar.2023.175517] [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: 10/17/2022] [Revised: 12/24/2022] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
BACKGROUND Apoptosis, oxidative stress, and neuroinflammation have been linked to the onset of Parkinson's disease (PD). Although the pre-treatment effects of Silibinin on a PD model have been evaluated, in the current study we investigated the chronic therapeutic effects of Silibinin microinjection on a rat model of established parkinsonism along with behavioral and laboratory markers assessments. METHOD Parkinsonism was induced by 6-hydroxydopamine (6-OHDA, 8 μg/2μl/rat). 21 days after that, animals were treated with Silibinin (100, 200, and 300 mg/kg for 15 consecutive days). Every two days, the bar test was used to evaluate Silibinin's anti-cataleptic properties. At the end, myeloperoxidase (MPO) activity and toll-like receptor 4 (TLR4) expression in the substantia nigra pars compacta (SNc), along with cerebrospinal fluid (CSF) levels of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, IL-6, caspase-3, Bax and Bcl-2 levels were assessed. We used homology modeling to predict the 3D structure of TLR4. RESULT Silibinin's Chronic treatment, dose-dependently decreased catalepsy. MPO activity and levels of TNF-α, IL-6, and IL-1β were reduced in Silibinin-treated rats in all three doses. Silibinin decreased Bax/Bcl-2 ratio, caspase-3, and downregulated TLR4 expression. Molecular docking revealed that there were hydrophobic and hydrogen bond interactions between the studied ligand and TLR4. Silibinin formed a stable complex with both monomer and dimer forms of TLR4. CONCLUSION In accordance with molecular modeling and alleviation of TLR4 activity with a consequent reduction in oxidative stress, restoration of CSF inflammatory cytokine, and minimization of SNc neuronal apoptosis, long-term therapy with Silibinin offers a potential opportunity for symptomatic PD treatment.
Collapse
Affiliation(s)
| | - Mohammadmahdi Sabahi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Seyed Ahmad Ebadi
- Department of Medicinal Chemistry, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zohre Sadeghian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Mohajjel Nayebi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rasool Haddadi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
14
|
Ni R. PET imaging in animal models of Parkinson's disease. Behav Brain Res 2023; 438:114174. [PMID: 36283568 DOI: 10.1016/j.bbr.2022.114174] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022]
Abstract
Alpha-synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies and multiple system atrophy, are characterized by aberrant accumulation of alpha-synuclein and synaptic dysfunction leading to motor and cognitive deficits. Animal models of alpha-synucleinopathy have greatly facilitated the mechanistic understanding of the disease and the development of therapeutics. Various transgenic, alpha-synuclein fibril-injected, and toxin-injected animal models of Parkinson's disease and multiple system atrophy that recapitulate the disease pathology have been developed and widely used. Recent advances in positron emission tomography have allowed the noninvasive visualization of molecular alterations, underpinning behavioral dysfunctions in the brains of animal models and the longitudinal monitoring of treatment effects. Imaging studies in these disease animal models have employed multi-tracer PET designs to reveal dopaminergic deficits together with other molecular alterations. This review focuses on the development of new positron emission tomography tracers and studies of alpha-synuclein, synaptic vesicle glycoprotein 2A neurotransmitter receptor deficits such as dopaminergic receptor, dopaminergic transporter, serotonergic receptor, vesicular monoamine transporter 2, hypometabolism, neuroinflammation, mitochondrial dysfunction and leucine rich repeat kinase 2 in animal models of Parkinson's disease. The outstanding challenges and emerging applications are outlined, such as investigating the gut-brain-axis by using positron emission tomography in animal models, and provide a future outlook.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
15
|
Qiu J, Peng G, Tang Y, Li S, Liu Z, Zheng J, Wang Y, Liu H, Wei L, Su Y, Lin Y, Dai W, Zhang Z, Chen X, Ding L, Guo W, Zhu X, Xu P, Mo M. Lipid profiles in the cerebrospinal fluid of rats with 6-hydroxydopamine-induced lesions as a model of Parkinson's disease. Front Aging Neurosci 2023; 14:1077738. [PMID: 36742201 PMCID: PMC9895836 DOI: 10.3389/fnagi.2022.1077738] [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: 10/23/2022] [Accepted: 12/30/2022] [Indexed: 01/21/2023] Open
Abstract
Background Parkinson's disease (PD) is a progressive neurodegenerative disease with characteristic pathological abnormalities, including the loss of dopaminergic (DA) neurons, a dopamine-depleted striatum, and microglial activation. Lipid accumulation exhibits a close relationship with these pathologies in PD. Methods Here, 6-hydroxydopamine (6-OHDA) was used to construct a rat model of PD, and the lipid profile in cerebrospinal fluid (CSF) obtained from model rats was analyzed using lipidomic approaches. Results Establishment of this PD model was confirmed by apomorphine-induced rotation behaviors, loss of DA neurons, depletion of dopamine in the striatum, and microglial activation after 6-OHDA-induced lesion generation. Unsupervised and supervised methods were employed for lipid analysis. A total of 172 lipid species were identified in CSF and subsequently classified into 18 lipid families. Lipid families, including eicosanoids, triglyceride (TG), cholesterol ester (CE), and free fatty acid (FFA), and 11 lipid species exhibited significantly altered profiles 2 weeks after 6-OHDA administration, and significant changes in eicosanoids, TG, CE, CAR, and three lipid species were noted 5 weeks after 6-OHDA administration. During the period of 6-OHDA-induced lesion formation, the lipid families and species showed concentration fluctuations related to the recovery of behavior and nigrostriatal abnormalities. Correlation analysis showed that the levels of eicosanoids, CE, TG families, and TG (16:0_20:0_18:1) exhibited positive relationships with apomorphine-induced rotation behaviors and negative relationships with tyrosine hydroxylase (TH) expression in the midbrain. Conclusion These results revealed that non-progressive nigrostriatal degeneration induced by 6-OHDA promotes the expression of an impairment-related lipidomic signature in CSF, and the level of eicosanoids, CE, TG families, and TG (16:0_20:0_18:1) in CSF may reveal pathological changes in the midbrain after 6-OHDA insult.
Collapse
Affiliation(s)
- Jiewen Qiu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoyou Peng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuting Tang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shiyin Li
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zengfu Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiayun Zheng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yunxin Wang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hanqun Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijian Wei
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yilin Su
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuwan Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Dai
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiling Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liuyan Ding
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenyuan Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoqin Zhu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingshu Mo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Internal Medicine, Huilai People’s Hospital, Jieyang, China
| |
Collapse
|
16
|
Sancandi M, De Caro C, Cypaite N, Marascio N, Avagliano C, De Marco C, Russo E, Constanti A, Mercer A. Effects of a probiotic suspension Symprove™ on a rat early-stage Parkinson's disease model. Front Aging Neurosci 2023; 14:986127. [PMID: 36742204 PMCID: PMC9890174 DOI: 10.3389/fnagi.2022.986127] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
An increasing number of studies in recent years have focused on the role that the gut may play in Parkinson's Disease (PD) pathogenesis, suggesting that the maintenance of a healthy gut may lead to potential treatments of the disease. The health of microbiota has been shown to be directly associated with parameters that play a potential role in PD including gut barrier integrity, immunity, function, metabolism and the correct functioning of the gut-brain axis. The gut microbiota (GM) may therefore be employed as valuable indicators for early diagnosis of PD and potential targets for preventing or treating PD symptoms. Preserving the gut homeostasis using probiotics may therefore lead to a promising treatment strategy due to their known benefits in improving constipation, motor impairments, inflammation, and neurodegeneration. However, the mechanisms underlying the effects of probiotics in PD are yet to be clarified. In this project, we have tested the efficacy of an oral probiotic suspension, Symprove™, on an established animal model of PD. Symprove™, unlike many commercially available probiotics, has been shown to be resistant to gastric acidity, improve symptoms in gastrointestinal diseases and improve gut integrity in an in vitro PD model. In this study, we used an early-stage PD rat model to determine the effect of Symprove™ on neurodegeneration and neuroinflammation in the brain and on plasma cytokine levels, GM composition and short chain fatty acid (SCFA) release. Symprove™ was shown to significantly influence both the gut and brain of the PD model. It preserved the gut integrity in the PD model, reduced plasma inflammatory markers and changed microbiota composition. The treatment also prevented the reduction in SCFAs and striatal inflammation and prevented tyrosine hydroxylase (TH)-positive cell loss by 17% compared to that observed in animals treated with placebo. We conclude that Symprove™ treatment may have a positive influence on the symptomology of early-stage PD with obvious implications for the improvement of gut integrity and possibly delaying/preventing the onset of neuroinflammation and neurodegeneration in human PD patients.
Collapse
Affiliation(s)
- Marco Sancandi
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom
| | - Carmen De Caro
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
| | - Neringa Cypaite
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom
| | - Nadia Marascio
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Carmela De Marco
- Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Emilio Russo
- Department of Science of Health, School of Medicine, University of Catanzaro, Catanzaro, Italy
| | - Andrew Constanti
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom
| | - Audrey Mercer
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom,*Correspondence: Audrey Mercer,
| |
Collapse
|
17
|
Advances in NURR1-Regulated Neuroinflammation Associated with Parkinson's Disease. Int J Mol Sci 2022; 23:ijms232416184. [PMID: 36555826 PMCID: PMC9788636 DOI: 10.3390/ijms232416184] [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: 10/24/2022] [Revised: 12/02/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Neuroinflammation plays a crucial role in the progression of neurodegenerative disorders, particularly Parkinson's disease (PD). Glial cell activation and subsequent adaptive immune involvement are neuroinflammatory features in familial and idiopathic PD, resulting in the death of dopaminergic neuron cells. An oxidative stress response, inflammatory mediator production, and immune cell recruitment and activation are all hallmarks of this activation, leading to chronic neuroinflammation and progressive neurodegeneration. Several studies in PD patients' cerebrospinal fluid and peripheral blood revealed alterations in inflammatory markers and immune cell populations that may lead to or exacerbate neuroinflammation and perpetuate the neurodegenerative process. Most of the genes causing PD are also expressed in astrocytes and microglia, converting their neuroprotective role into a pathogenic one and contributing to disease onset and progression. Nuclear receptor-related transcription factor 1 (NURR1) regulates gene expression linked to dopaminergic neuron genesis and functional maintenance. In addition to playing a key role in developing and maintaining neurotransmitter phenotypes in dopaminergic neurons, NURR1 agonists have been shown to reverse behavioral and histological abnormalities in animal PD models. NURR1 protects dopaminergic neurons from inflammation-induced degeneration, specifically attenuating neuronal death by suppressing the expression of inflammatory genes in microglia and astrocytes. This narrative review highlights the inflammatory changes in PD and the advances in NURR1-regulated neuroinflammation associated with PD. Further, we present new evidence that targeting this inflammation with a variety of potential NURR1 target therapy medications can effectively slow the progression of chronic neuroinflammation-induced PD.
Collapse
|
18
|
Chronic Treatment with the Probiotics Lacticaseibacillus rhamnosus GG and Bifidobacterium lactis BB12 Attenuates Motor Impairment, Striatal Microglial Activation, and Dopaminergic Loss in Rats with 6-Hydroxydopamine-induced Hemiparkinsonism. Neuroscience 2022; 507:79-98. [PMID: 36370934 DOI: 10.1016/j.neuroscience.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/05/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Gut dysbiosis is considered a risk factor for Parkinson's disease (PD), and chronic treatment with probiotics could prevent it. Here we report the assessment of a probiotic mixture [Lacticaseibacillus rhamnosus GG (LGG), and Bifidobacterium animalis lactis BB-12 (BB-12)] administered to male rats 2 weeks before and 3 weeks after injecting 6-hydroxydopamine (6-OHDA) into the right striatum, a model that mimics the early stages of PD. Before and after lesion, animals were subjected to behavioral tests: narrow beam, cylinder test, and apomorphine (APO)-induced rotations. Dopaminergic (DA) denervation and microglia recruitment were assessed with tyrosine hydroxylase (TH+) and ionized calcium-binding protein-1 adapter (Iba1+) immunostaining, respectively. Post 6-OHDA injury, rats treated with sunflower oil (probiotics vehicle) developed significant decrease in crossing speed and increases in contralateral paw slips (narrow beam), forepaw use asymmetry (cylinder), and APO-induced rotations. In striatum, 6-OHDA eliminated ≈2/3 of TH+ area and caused significant increase of Iba1+ microglia population. Retrograde axonal degeneration suppressed ≈2/5 of TH+ neurons in the substantia nigra pars compacta (SNpc). In hemiparkinsonian rats, probiotics treatment significantly improved the crossing speed, and also reduced paw slips (postlesion days 14 and 21), the loss of TH+ neurons in SNpc, and the loss of TH+ area and of Iba1+ microglia count in striatum, without affecting the proportion of microglia morphological phenotypes. Probiotics treatment did not attenuate forepaw use asymmetry nor APO-induced rotations. These results indicate that the mixture of probiotics LGG and BB-12 protects nigrostriatal DA neurons against 6-OHDA-induced damage, supporting their potential as preventive treatment of PD.
Collapse
|
19
|
Zhang C, Shi G, Li G, Zuo K, Bai X, Meng Q, Huang M. Paraquat induces microglial cause early neuronal synaptic deficits through activation of the classical complement cascade response. Immunobiology 2022; 227:152275. [PMID: 36152426 DOI: 10.1016/j.imbio.2022.152275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/30/2022] [Accepted: 09/01/2022] [Indexed: 11/05/2022]
Abstract
Synapse loss is considered to be an early event in the dysfunction of the central nervous system (CNS), precedes neuronal decline, which is the main pathological change in mild cognitive impairment (MCI). Accumulating evidence has shown that neuronal synapse loss is associated with hyperactivity of microglial phagocytosis. In the CNS, microglia act as macrophages to clear cellular debris and weakened synapses, but the mechanism by which microglia activation leads to phagocytosis disorder remains unclear. Therefore, we treated mice with paraquat (PQ) in intraperitoneal injection to explore the mechanism by which microglia exert immunotoxicity in the CNS and cause neuronal synapse loss. Immunofluorescence results exposed synapses decreased with PQ exposure time, but the staining HE and Nissl showed that neuronal cell bodies were hardly affected. Fluorescence co-localization found that C1q (classical complement cascade initiation factor) was gradually deposited in the postsynaptic membrane of neurons to trigger the cascade reaction, thereby causing the excessive deposition of C3, a key factor of the classical complement cascade, and further induces hyperactivation of microglia, leads to phagocytosis disorder. IHC results demonstrated that the parallel relationship. Therefore, all our preliminary results throw light on the mechanism by which PQ abnormally triggers the immune system to produce immunotoxicity leading to microglial phagocytic dysfunction.
Collapse
Affiliation(s)
- Chunhui Zhang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Ge Shi
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Guoliang Li
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Kai Zuo
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Xinghua Bai
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Qi Meng
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China
| | - Min Huang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan, China.
| |
Collapse
|
20
|
Leveraging the preformed fibril model to distinguish between alpha-synuclein inclusion- and nigrostriatal degeneration-associated immunogenicity. Neurobiol Dis 2022; 171:105804. [PMID: 35764290 PMCID: PMC9803935 DOI: 10.1016/j.nbd.2022.105804] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/04/2022] [Accepted: 06/22/2022] [Indexed: 01/03/2023] Open
Abstract
Neuroinflammation has become a well-accepted pathologic hallmark of Parkinson's disease (PD). However, it remains unclear whether inflammation, triggered by α-syn aggregation and/or degeneration, contributes to the progression of the disease. Studies examining neuroinflammation in PD are unable to distinguish between Lewy body-associated inflammation and degeneration-associated inflammation, as both pathologies are present simultaneously. Intrastriatal and intranigral injections of alpha-synuclein (α-syn) preformed fibrils (PFFs) results in two distinct pathologic phases: Phase 1: The accumulation and peak formation of α-syn inclusions in nigrostriatal system and, Phase 2: Protracted dopaminergic neuron degeneration. In this review we summarize the current understanding of neuroinflammation in the α-syn PFF model, leveraging the distinct Phase 1 aggregation phase and Phase 2 degeneration phase to guide our interpretations. Studies consistently demonstrate an association between pathologic α-syn aggregation in the substantia nigra (SN) and activation of the innate immune system. Further, major histocompatibility complex-II (MHC-II) antigen presentation is proportionate to inclusion load. The α-syn aggregation phase is also associated with peripheral and adaptive immune cell infiltration to the SN. These findings suggest that α-syn like aggregates are immunogenic and thus have the potential to contribute to the degenerative process. Studies examining neuroinflammation during the neurodegenerative phase reveal elevated innate, adaptive, and peripheral immune cell markers, however limitations of single time point experimental design hinder interpretations as to whether this neuroinflammation preceded, or was triggered by, nigral degeneration. Longitudinal studies across both the aggregation and degeneration phases of the model suggest that microglial activation (MHC-II) is greater in magnitude during the aggregation phase that precedes degeneration. Overall, the consistency between neuroinflammatory markers in the parkinsonian brain and in the α-syn PFF model, combined with the distinct aggregation and degenerative phases, establishes the utility of this model platform to yield insights into pathologic events that contribute to neuroinflammation and disease progression in PD.
Collapse
|
21
|
Weiss F, Labrador-Garrido A, Dzamko N, Halliday G. Immune responses in the Parkrtdinson's disease brain. Neurobiol Dis 2022; 168:105700. [DOI: 10.1016/j.nbd.2022.105700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/15/2022] Open
|
22
|
Quijano A, Diaz-Ruiz C, Lopez-Lopez A, Villar-Cheda B, Muñoz A, Rodriguez-Perez AI, Labandeira-Garcia JL. Angiotensin Type-1 Receptor Inhibition Reduces NLRP3 Inflammasome Upregulation Induced by Aging and Neurodegeneration in the Substantia Nigra of Male Rodents and Primary Mesencephalic Cultures. Antioxidants (Basel) 2022; 11:antiox11020329. [PMID: 35204211 PMCID: PMC8868290 DOI: 10.3390/antiox11020329] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/31/2022] [Accepted: 02/05/2022] [Indexed: 12/17/2022] Open
Abstract
The tissue renin–angiotensin system (RAS) has been shown to be involved in prooxidative and proinflammatory changes observed in aging and aging-related diseases such as dopaminergic degeneration in Parkinson’s disease (PD). We studied the activation of the NLRP3 inflammasome in the substantia nigra with aging and early stages of dopaminergic degeneration in PD models and, particularly, if the brain RAS, via its prooxidative proinflammatory angiotensin II (AngII) type 1 (AT1) receptors, mediates the inflammasome activation. Nigras from aged rats and mice and 6-hydroxydopamine PD models showed upregulation in transcription of inflammasome-related components (NLRP3, pro-IL1β and pro-IL18) and IL1β and IL18 protein levels, which was inhibited by the AT1 receptor antagonist candesartan. The role of the AngII/AT1 axis in inflammasome activation was further confirmed in rats intraventricularly injected with AngII, and in primary mesencephalic cultures treated with 6-hydroxydopamine, which showed inflammasome activation that was blocked by candesartan. Observations in the nigra of young and aged AT1 and AT2 knockout mice confirmed the major role of AT1 receptors in nigral inflammasome activation. In conclusion, the inflammasome is upregulated by aging and dopaminergic degeneration in the substantia nigra, possibly related with a decrease in dopamine levels, and it is mediated by the AngII/AT1 axis.
Collapse
Affiliation(s)
- Aloia Quijano
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
| | - Carmen Diaz-Ruiz
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Andrea Lopez-Lopez
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Begoña Villar-Cheda
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Ana Muñoz
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Ana I. Rodriguez-Perez
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Jose L. Labandeira-Garcia
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
- Correspondence: ; Tel.: +34-881-812223
| |
Collapse
|
23
|
Banerjee R, Rai A, Iyer SM, Narwal S, Tare M. Animal models in the study of Alzheimer's disease and Parkinson's disease: A historical perspective. Animal Model Exp Med 2022; 5:27-37. [PMID: 35229999 PMCID: PMC8879627 DOI: 10.1002/ame2.12209] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/20/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease and Parkinson's disease are two of the most prevalent and disabling neurodegenerative diseases globally. Both are proteinopathic conditions and while occasionally inherited, are largely sporadic in nature. Although the advances in our understanding of the two have been significant, they are far from complete and neither diagnosis nor the current practices in treatment and rehabilitation is adequately helpful. Animal models have historically found application as testing beds for novel therapeutics and continue to be valuable aids in pharmacological research. This review chronicles the development of those models in the context of Alzheimer's and Parkinson's disease, and highlights the shifting paradigms in studying two human-specific conditions in non-human organisms.
Collapse
Affiliation(s)
- Rajashree Banerjee
- Department of Biological SciencesBirla Institute of Technology and SciencePilaniIndia
| | - Arushi Rai
- Department of Biological SciencesBirla Institute of Technology and SciencePilaniIndia
| | - Shreyas M. Iyer
- Department of Biological SciencesBirla Institute of Technology and SciencePilaniIndia
| | - Sonia Narwal
- Department of Biological SciencesBirla Institute of Technology and SciencePilaniIndia
| | - Meghana Tare
- Department of Biological SciencesBirla Institute of Technology and SciencePilaniIndia
| |
Collapse
|
24
|
Saha S, Buttari B, Profumo E, Tucci P, Saso L. A Perspective on Nrf2 Signaling Pathway for Neuroinflammation: A Potential Therapeutic Target in Alzheimer's and Parkinson's Diseases. Front Cell Neurosci 2022; 15:787258. [PMID: 35126058 PMCID: PMC8813964 DOI: 10.3389/fncel.2021.787258] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation plays a pivotal role in Alzheimer's disease (AD) and Parkinson's disease (PD), the leading causes of dementia. These neurological disorders are characterized by the accumulation of misfolded proteins such as amyloid-ß (Aß), tau protein and α-synuclein, contributing to mitochondrial fragmentation, oxidative stress, and neuroinflammation. Misfolded proteins activate microglia, which induces neuroinflammation, expression of pro-inflammatory cytokines and subsequently facilitates synaptic damage and neuronal loss. So far, all the proposed drugs were based on the inhibition of protein aggregation and were failed in clinical trials. Therefore, the treatment options of dementia are still a challenging issue. Thus, it is worthwhile to study alternative therapeutic strategies. In this context, there is increasing data on the pivotal role of transcription factor NF- E2 p45-related factor 2 (Nrf2) on the redox homeostasis and anti-inflammatory functions in neurodegenerative disorders. Interestingly, Nrf2 signaling pathway has shown upregulation of antioxidant genes, inhibition of microglia-mediated inflammation, and improved mitochondrial function in neurodegenerative diseases, suggesting Nrf2 activation could be a novel therapeutic approach to target pathogenesis. The present review will examine the correlation between Nrf2 signaling with neuroinflammation in AD and PD.
Collapse
Affiliation(s)
- Sarmistha Saha
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Italian National Institute of Health, Rome, Italy
| | - Brigitta Buttari
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Italian National Institute of Health, Rome, Italy
| | - Elisabetta Profumo
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Italian National Institute of Health, Rome, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Piri H, Sharifi S, Nigjeh S, Haghdoost-Yazdi H. Dopaminergic neuronal death in the substantia nigra associates with change in serum levels of TNF-α and IL-1β; evidence from early experimental model of Parkinson's disease. Neurol Res 2022; 44:544-553. [PMID: 34986749 DOI: 10.1080/01616412.2021.2024726] [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] [Indexed: 10/19/2022]
Abstract
PURPOSE Studies have shown that inflammation plays a key role in etiology of Parkinson's disease (PD). However, human studies which have evaluated association between PD and serum levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) have reported conflicting results. In this study, serum and striatum levels of these cytokines were evaluated in 6-hydroxydopamine (6-OHDA) animal model of PD. METHOD The neurotoxin of 6-OHDA was injected into medial forebrain bundle of right hemisphere and behavioral tests were carried out to eight weeks thereafter to evaluate severity of PD and its progress. Blood was collected before the toxin and in second and eight weeks after that. Survival of dopaminergic (DAergic) neurons in substantia nigra was assessed by immunohistochemistry. TNF-α and IL-1β levels were determined using ELISA kits. RESULT Severity of behavioral symptoms was gradually increased in 6-OHDA-treated rats. They showed a decrease in serum TNF-α level in the eight week and increase in IL-1β both in the second and eight weeks. They were divided into two subgroups, symptomatic and asymptomatic with severe and moderate degrees in DAergic neuronal death. Significant decrease in serum TNF-α was only observed in the symptomatic subgroup but IL-1β increased in both subgroups. Also, striatal levels of both cytokines were higher in the lesioned hemisphere. CONCLUSION Increase in serum IL-1β level can reflect moderate degree of lesion in substantia nigra and thereby is used for prognosis of PD before its clinical symptoms are appeared. On the other hand, an increase in serum TNF-α is appeared in advanced stage of PD.
Collapse
Affiliation(s)
- Hossein Piri
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sahar Sharifi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sepideh Nigjeh
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hashem Haghdoost-Yazdi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
26
|
Endepols H, Zlatopolskiy BD, Zischler J, Alavinejad N, Apetz N, Vus S, Drzezga A, Neumaier B. Imaging of cerebral tryptophan metabolism using 7-[ 18F]FTrp-PET in a unilateral Parkinsonian rat model. Neuroimage 2021; 247:118842. [PMID: 34942366 DOI: 10.1016/j.neuroimage.2021.118842] [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: 07/30/2021] [Revised: 11/30/2021] [Accepted: 12/19/2021] [Indexed: 10/19/2022] Open
Abstract
Degradation products of the essential amino acid tryptophan (Trp) are important signaling molecules in the mammalian brain. Trp is metabolized either through the kynurenine pathway or enters serotonin and melatonin syntheses. The aim of the present work was to examine the potential of the novel PET tracer 7-[18F]fluorotryptophan ([18F]FTrp) to visualize all three pathways in a unilateral 6-OHDA rat model. [18F]FDOPA-PET scans were performed in nine 6-OHDA-injected and six sham-operated rats to assess unilateral dopamine depletion severity four weeks after lesion placement. Afterwards, 7-[18F]FTrp-PET scans were conducted at different timepoints up to seven months after 6-OHDA injection. In addition, two 6-OHDA-injected rats were examined for neuroinflammation using [18F]DAA1106-PET. 7-[18F]FTrp-PET showed significantly increased tracer uptake at the 6-OHDA injection site which was negatively correlated to time after lesion placement. Accumulation of [18F]DAA1106 at the injection site was increased as well, suggesting that 7-[18F]FTrp uptake in this region may reflect kynurenine pathway activity associated with inflammation. Bilaterally in the dorsal hippocampus, 7-[18F]FTrp uptake was significantly decreased and was inversely correlated to dopamine depletion severity, indicating that it reflects reduced serotonin synthesis. Finally, 7-[18F]FTrp uptake in the pineal gland was significantly increased in relation with dopamine depletion severity, providing evidence that melatonin synthesis is increased in the 6-OHDA rat model. We conclude that 7-[18F]FTrp is able to detect alterations in both serotonin/melatonin and kynurenine metabolic pathways, and can be applied to visualize pathologic changes related to neurodegenerative processes.
Collapse
Affiliation(s)
- Heike Endepols
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Kerpener Str. 62, 50937 Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, Kerpener Str. 62, 50937 Cologne, Germany; Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52425 Jülich, Germany.
| | - Boris D Zlatopolskiy
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Kerpener Str. 62, 50937 Cologne, Germany; Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52425 Jülich, Germany.
| | - Johannes Zischler
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Kerpener Str. 62, 50937 Cologne, Germany; Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52425 Jülich, Germany
| | - Nazanin Alavinejad
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Kerpener Str. 62, 50937 Cologne, Germany.
| | - Nadine Apetz
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Kerpener Str. 62, 50937 Cologne, Germany.
| | - Stefanie Vus
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Kerpener Str. 62, 50937 Cologne, Germany; Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52425 Jülich, Germany.
| | - Alexander Drzezga
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Nuclear Medicine, Kerpener Str. 62, 50937 Cologne, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany; Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Molecular Organization of the Brain (INM-2), Wilhelm-Johnen-Straße, 52425 Jülich, Germany.
| | - Bernd Neumaier
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, Kerpener Str. 62, 50937 Cologne, Germany; Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52425 Jülich, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany.
| |
Collapse
|
27
|
Van Camp N, Lavisse S, Roost P, Gubinelli F, Hillmer A, Boutin H. TSPO imaging in animal models of brain diseases. Eur J Nucl Med Mol Imaging 2021; 49:77-109. [PMID: 34245328 PMCID: PMC8712305 DOI: 10.1007/s00259-021-05379-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/25/2021] [Indexed: 12/19/2022]
Abstract
Over the last 30 years, the 18-kDa TSPO protein has been considered as the PET imaging biomarker of reference to measure increased neuroinflammation. Generally assumed to image activated microglia, TSPO has also been detected in endothelial cells and activated astrocytes. Here, we provide an exhaustive overview of the recent literature on the TSPO-PET imaging (i) in the search and development of new TSPO tracers and (ii) in the understanding of acute and chronic neuroinflammation in animal models of neurological disorders. Generally, studies testing new TSPO radiotracers against the prototypic [11C]-R-PK11195 or more recent competitors use models of acute focal neuroinflammation (e.g. stroke or lipopolysaccharide injection). These studies have led to the development of over 60 new tracers during the last 15 years. These studies highlighted that interpretation of TSPO-PET is easier in acute models of focal lesions, whereas in chronic models with lower or diffuse microglial activation, such as models of Alzheimer's disease or Parkinson's disease, TSPO quantification for detection of neuroinflammation is more challenging, mirroring what is observed in clinic. Moreover, technical limitations of preclinical scanners provide a drawback when studying modest neuroinflammation in small brains (e.g. in mice). Overall, this review underlines the value of TSPO imaging to study the time course or response to treatment of neuroinflammation in acute or chronic models of diseases. As such, TSPO remains the gold standard biomarker reference for neuroinflammation, waiting for new radioligands for other, more specific targets for neuroinflammatory processes and/or immune cells to emerge.
Collapse
Affiliation(s)
- Nadja Van Camp
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Sonia Lavisse
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Pauline Roost
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Francesco Gubinelli
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Ansel Hillmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, CT, USA
| | - Hervé Boutin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Brain and Mental Health, University of Manchester, M13 9PL, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, 27 Palatine Road, M20 3LJ, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
28
|
Intranasal Exposure to Low-Dose Rotenone Induced Alpha-Synuclein Accumulation and Parkinson's Like Symptoms Without Loss of Dopaminergic Neurons. Neurotox Res 2021; 40:215-229. [PMID: 34817799 DOI: 10.1007/s12640-021-00436-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/21/2022]
Abstract
Epidemiologically Parkinson's disease (PD) is associated with chronic ingestion or inhalation of environmental toxins leading to the development of motor symptoms. Though neurotoxin-based animal models played a major role in understanding diverse pathogenesis, they failed to identify the risk assessment due to uncommon route of toxin exposure. Towards this, the available neurotoxin-based intranasal (i.n.) PD models targeting olfactory bulb (OB) have demonstrated the dopaminergic (DAergic) neurodegeneration in both OB and substantia nigra (SN). Despite that, the studies detecting the alpha-synuclein (α-syn) accumulation in OB and its progression to other brain regions due to inhalation of environmental toxins are still lacking. Herein, we developed oil in water microemulsion of rotenone administered intranasally to the mice at a dose which is not detectable in blood, brain, and olfactory bulb by LCMS method. Our data reveals that 9 weeks of rotenone exposure did not induce olfactory and motor dysfunction. Conversely, after 16 weeks of washout period, rotenone treated mice showed both olfactory and motor impairment, along with α-syn accumulation in the OB and striatum without glial cell activation and loss of dopaminergic neurons. The results depict the progressive nature of the developed model and highlight the role of α-syn in PD like pathology or symptoms. Together, our findings suggest the adverse consequences of early exposure to the environmental toxins on the olfactory system for a shorter period with relevance to the development of synucleinopathy or Parkinson's disease in its later stage.
Collapse
|
29
|
Alagaratnam J, Winston A. Molecular neuroimaging of inflammation in HIV. Clin Exp Immunol 2021; 210:14-23. [PMID: 35020855 PMCID: PMC9585552 DOI: 10.1093/cei/uxab013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/13/2021] [Accepted: 11/03/2021] [Indexed: 01/12/2023] Open
Abstract
People with HIV now have near-normal life expectancies due to the success of effective combination antiretroviral therapy (cART). Following cART initiation, immune recovery occurs, and opportunistic diseases become rare. Despite this, high rates of non-infectious comorbidities persist in treated people with HIV, hypothesized to be related to persistent immuno-activation. One such comorbidity is cognitive impairment, which may partly be driven by ongoing neuro-inflammation in otherwise effectively treated people with HIV. In order to develop therapeutic interventions to address neuro-inflammation in effectively treated people with HIV, a deeper understanding of the pathogenic mechanisms driving persistent neuro-inflammatory responses and the ability to better characterize and measure neuro-inflammation in the central nervous system is required. This review highlights recent advances in molecular neuroimaging techniques which have the potential to assess neuro-inflammatory responses within the central nervous system in HIV disease. Proton magnetic resonance spectroscopy (1H-MRS) has been utilized to assess neuro-inflammatory responses since early in the HIV pandemic and shows promise in recent studies assessing different antiretroviral regimens. 1H-MRS is widely available in both resource-rich and some resource-constrained settings and is relatively inexpensive. Brain positron emission tomography (PET) imaging using Translocator Protein (TSPO) radioligands is a rapidly evolving field; newer TSPO-radioligands have lower signal-to-noise ratio and have the potential to localize neuro-inflammation within the brain in people with HIV. As HIV therapeutics evolve, people with HIV continue to age and develop age-related comorbidities including cognitive disorders. The use of novel neuroimaging modalities in the field is likely to advance in order to rapidly assess novel therapeutic interventions and may play a role in future clinical assessments.
Collapse
Affiliation(s)
- Jasmini Alagaratnam
- Correspondence: Jasmini Alagaratnam, Clinical Trials Centre, Winston Churchill Wing, St. Mary’s Hospital, Praed Street, London W2 1NY, UK.
| | - Alan Winston
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK,Department of Genitourinary Medicine & HIV, St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
30
|
Bido S, Muggeo S, Massimino L, Marzi MJ, Giannelli SG, Melacini E, Nannoni M, Gambarè D, Bellini E, Ordazzo G, Rossi G, Maffezzini C, Iannelli A, Luoni M, Bacigaluppi M, Gregori S, Nicassio F, Broccoli V. Microglia-specific overexpression of α-synuclein leads to severe dopaminergic neurodegeneration by phagocytic exhaustion and oxidative toxicity. Nat Commun 2021; 12:6237. [PMID: 34716339 PMCID: PMC8556263 DOI: 10.1038/s41467-021-26519-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Recent findings in human samples and animal models support the involvement of inflammation in the development of Parkinson's disease. Nevertheless, it is currently unknown whether microglial activation constitutes a primary event in neurodegeneration. We generated a new mouse model by lentiviral-mediated selective α-synuclein (αSYN) accumulation in microglial cells. Surprisingly, these mice developed progressive degeneration of dopaminergic (DA) neurons without endogenous αSYN aggregation. Transcriptomics and functional assessment revealed that αSYN-accumulating microglial cells developed a strong reactive state with phagocytic exhaustion and excessive production of oxidative and proinflammatory molecules. This inflammatory state created a molecular feed-forward vicious cycle between microglia and IFNγ-secreting immune cells infiltrating the brain parenchyma. Pharmacological inhibition of oxidative and nitrosative molecule production was sufficient to attenuate neurodegeneration. These results suggest that αSYN accumulation in microglia induces selective DA neuronal degeneration by promoting phagocytic exhaustion, an excessively toxic environment and the selective recruitment of peripheral immune cells.
Collapse
Affiliation(s)
- Simone Bido
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Sharon Muggeo
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Luca Massimino
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Matteo Jacopo Marzi
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Serena Gea Giannelli
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Elena Melacini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- National Research Council (CNR), Institute of Neuroscience, 20129, Milan, Italy
| | - Melania Nannoni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Diana Gambarè
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Edoardo Bellini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Gabriele Ordazzo
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Greta Rossi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Camilla Maffezzini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Angelo Iannelli
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- National Research Council (CNR), Institute of Neuroscience, 20129, Milan, Italy
| | - Mirko Luoni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Marco Bacigaluppi
- Division of Neuroscience, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Nicassio
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), 20139, Milan, Italy
| | - Vania Broccoli
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
- National Research Council (CNR), Institute of Neuroscience, 20129, Milan, Italy.
| |
Collapse
|
31
|
Guo M, Schwartz TD, Dunaief JL, Cui QN. Myeloid cells in retinal and brain degeneration. FEBS J 2021; 289:2337-2361. [PMID: 34478598 PMCID: PMC8891394 DOI: 10.1111/febs.16177] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/06/2021] [Accepted: 09/02/2021] [Indexed: 12/11/2022]
Abstract
Retinal inflammation underlies multiple prevalent ocular and neurological diseases. Similar inflammatory processes are observed in glaucomatous optic neuropathy, age-related macular degeneration, retinitis pigmentosa, posterior uveitis, Alzheimer's disease, and Parkinson's disease. In particular, human and animal studies have demonstrated the important role microglia/macrophages play in initiating and maintaining a pro-inflammatory environment in degenerative processes impacting vision. On the other hand, microglia have also been shown to have a protective role in multiple central nervous system diseases. Identifying the mechanisms underlying cell dysfunction and death is the first step toward developing novel therapeutics for these diseases impacting the central nervous system. In addition to reviewing recent key studies defining important mediators of retinal inflammation, with an emphasis on translational studies that bridge this research from bench to bedside, we also highlight a promising therapeutic class of medications, the glucagon-like peptide-1 receptor agonists. Finally, we propose areas where additional research is necessary to identify mechanisms that can be modulated to shift the balance from a neurotoxic to a neuroprotective retinal environment.
Collapse
Affiliation(s)
- Michelle Guo
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Turner D Schwartz
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua L Dunaief
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Qi N Cui
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
32
|
Quadir H, Hakobyan K, Gaddam M, Ojinnaka U, Ahmed Z, Kannan A, Mostafa JA. Role of Rho-Associated Protein Kinase Inhibition As Therapeutic Strategy for Parkinson's Disease: Dopaminergic Survival and Enhanced Mitophagy. Cureus 2021; 13:e16973. [PMID: 34377615 PMCID: PMC8349301 DOI: 10.7759/cureus.16973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/07/2021] [Indexed: 12/24/2022] Open
Abstract
The GTP-binding protein, Rho, plays a significant role in the cellular pathology of Parkinson’s disease. The downstream effector of Rho, Rho-associated kinase (ROCK), performs several functions, including microglial inflammatory response and enhanced Parkin-mediated mitophagy. Its inhibition shows neuroprotective effects in carried studies. Parkinson’s disease pathology also rests on incomplete removal of damaged mitochondria, leading to neuronal impairment. ROCK has different isoforms, inhibition of which have been shown to decrease the adverse changes in microglia. There has also been evidence of a decreased release of inflammatory cytokines and a reduction in degradation of dopaminergic neurons on the addition of ROCK inhibitors. Additionally, ROCK inhibitors have recently been shown to increase the activity of hexokinase 2 (HK2), relocating it to mitochondria, and therefore leading to upregulated mitochondrial targeting. Understanding the cellular basis of ROCK activity and its inhibition may help us advance in creating new strategies for the treatment of Parkinson’s disease.
Collapse
Affiliation(s)
- Huma Quadir
- Internal Medicine/Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Knkush Hakobyan
- Diagnostic Radiology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mrunanjali Gaddam
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ugochi Ojinnaka
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Zubayer Ahmed
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Amudhan Kannan
- Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND.,General Surgery Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jihan A Mostafa
- Faculty Member, California Institute of Behavioral Neurosciences & Psychology, Fairfield, California, USA
| |
Collapse
|
33
|
Chen S, Annesley SJ, Jasim RAF, Fisher PR. The Parkinson's Disease-Associated Protein DJ-1 Protects Dictyostelium Cells from AMPK-Dependent Outcomes of Oxidative Stress. Cells 2021; 10:cells10081874. [PMID: 34440642 PMCID: PMC8392454 DOI: 10.3390/cells10081874] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/17/2021] [Accepted: 07/22/2021] [Indexed: 01/24/2023] Open
Abstract
Mitochondrial dysfunction has been implicated in the pathology of Parkinson’s disease (PD). In Dictyostelium discoideum, strains with mitochondrial dysfunction present consistent, AMPK-dependent phenotypes. This provides an opportunity to investigate if the loss of function of specific PD-associated genes produces cellular pathology by causing mitochondrial dysfunction with AMPK-mediated consequences. DJ-1 is a PD-associated, cytosolic protein with a conserved oxidizable cysteine residue that is important for the protein’s ability to protect cells from the pathological consequences of oxidative stress. Dictyostelium DJ-1 (encoded by the gene deeJ) is located in the cytosol from where it indirectly inhibits mitochondrial respiration and also exerts a positive, nonmitochondrial role in endocytosis (particularly phagocytosis). Its loss in unstressed cells impairs endocytosis and causes correspondingly slower growth, while also stimulating mitochondrial respiration. We report here that oxidative stress in Dictyostelium cells inhibits mitochondrial respiration and impairs phagocytosis in an AMPK-dependent manner. This adds to the separate impairment of phagocytosis caused by DJ-1 knockdown. Oxidative stress also combines with DJ-1 loss in an AMPK-dependent manner to impair or exacerbate defects in phototaxis, morphogenesis and growth. It thereby phenocopies mitochondrial dysfunction. These results support a model in which the oxidized but not the reduced form of DJ-1 inhibits AMPK in the cytosol, thereby protecting cells from the adverse consequences of oxidative stress, mitochondrial dysfunction and the resulting AMPK hyperactivity.
Collapse
Affiliation(s)
- Suwei Chen
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC 3086, Australia; (S.C.); (S.J.A.); (R.A.F.J.)
- School of Modern Agriculture and Biotechnology, Ankang University, Ankang 725000, China
| | - Sarah J. Annesley
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC 3086, Australia; (S.C.); (S.J.A.); (R.A.F.J.)
| | - Rasha A. F. Jasim
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC 3086, Australia; (S.C.); (S.J.A.); (R.A.F.J.)
- Department of Laboratory and Clinical Sciences, College of Pharmacy, University of Babylon, Hillah 51002, Iraq
| | - Paul R. Fisher
- Discipline of Microbiology, Department of Physiology Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC 3086, Australia; (S.C.); (S.J.A.); (R.A.F.J.)
- Correspondence: ; Tel.: +61-3-9479-2229
| |
Collapse
|
34
|
The impact of sesamol and exercise on striatal TNF-α level, motor behavior, aversive memory and oxidative stress status in 6-hydroxydopamine-lesioned rats. PHYSIOLOGY AND PHARMACOLOGY 2021. [DOI: 10.52547/phypha.26.1.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
35
|
Zheng T, Zhang Z. Activated microglia facilitate the transmission of α-synuclein in Parkinson's disease. Neurochem Int 2021; 148:105094. [PMID: 34097990 DOI: 10.1016/j.neuint.2021.105094] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/23/2021] [Accepted: 05/31/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta and abnormal aggregates of α-synuclein protein called Lewy bodies. To date, there is no drug that can definitely slow down or stop the progression of this disease. The discovery of the cell-to-cell transmission of pathologic α-synuclein seeds offers the possibility to explore novel treatment strategies to prevent the spread of α-synuclein, with the purpose of slowing down the progression of PD in its tracks. Although recent studies have made tremendous progress in understanding how α-synuclein spreads throughout the brain, neuroinflammation seems to play a crucial role in the development of α-synuclein pathology in PD. The activation of microglia, one of the hallmarks of the neuroinflammatory process, is suggested to influence the neuron-to-neuron transmission of α-synuclein. This review summarizes how activated microglia facilitate this process, and focuses on the following mechanisms including the activation of microglia in PD, the reduced ability of activated microglia to clear α-synuclein and increased migratory capacity of microglia in PD, as well as the cooperation between microglia and exosomes in mediating α-synuclein release and propagation. In conclusion, this article help collate information on microglia in-relation to PD.
Collapse
Affiliation(s)
- Tingting Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China
| | - Zhengxiang Zhang
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China.
| |
Collapse
|
36
|
Zaman V, Shields DC, Shams R, Drasites KP, Matzelle D, Haque A, Banik NL. Cellular and molecular pathophysiology in the progression of Parkinson's disease. Metab Brain Dis 2021; 36:815-827. [PMID: 33599945 PMCID: PMC8170715 DOI: 10.1007/s11011-021-00689-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder etiologically linked to the loss of substantia nigra (SN) dopaminergic neurons in the mid-brain. The etiopathology of sporadic PD is still unclear; however, the interaction of extrinsic and intrinsic factors may play a critical role in the onset and progression of the disease. Studies in animal models and human post-mortem tissue have identified distinct cellular and molecular changes in the diseased brain, suggesting complex interactions between different glial cell types and various molecular pathways. Small changes in the expression of specific genes in a single pathway or cell type possibly influence others at the cellular and system levels. These molecular and cellular signatures like neuroinflammation, oxidative stress, and autophagy have been observed in PD patients' brain tissue. While the etiopathology of PD is still poorly understood, the interplay between glial cells and molecular events may play a crucial role in disease onset and progression.
Collapse
Affiliation(s)
- Vandana Zaman
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
| | - Donald C Shields
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
| | - Ramsha Shams
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Kelsey P Drasites
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Denise Matzelle
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| | - Narendra L Banik
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA.
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA.
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| |
Collapse
|
37
|
El-Gamal M, Salama M, Collins-Praino LE, Baetu I, Fathalla AM, Soliman AM, Mohamed W, Moustafa AA. Neurotoxin-Induced Rodent Models of Parkinson's Disease: Benefits and Drawbacks. Neurotox Res 2021; 39:897-923. [PMID: 33765237 DOI: 10.1007/s12640-021-00356-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by cardinal motor impairments, including akinesia and tremor, as well as by a host of non-motor symptoms, including both autonomic and cognitive dysfunction. PD is associated with a death of nigral dopaminergic neurons, as well as the pathological spread of Lewy bodies, consisting predominantly of the misfolded protein alpha-synuclein. To date, only symptomatic treatments, such as levodopa, are available, and trials aiming to cure the disease, or at least halt its progression, have not been successful. Wong et al. (2019) suggested that the lack of effective therapy against neurodegeneration in PD might be attributed to the fact that the molecular mechanisms standing behind the dopaminergic neuronal vulnerability are still a major scientific challenge. Understanding these molecular mechanisms is critical for developing effective therapy. Thirty-five years ago, Calne and William Langston (1983) raised the question of whether biological or environmental factors precipitate the development of PD. In spite of great advances in technology and medicine, this question still lacks a clear answer. Only 5-15% of PD cases are attributed to a genetic mutation, with the majority of cases classified as idiopathic, which could be linked to exposure to environmental contaminants. Rodent models play a crucial role in understanding the risk factors and pathogenesis of PD. Additionally, well-validated rodent models are critical for driving the preclinical development of clinically translatable treatment options. In this review, we discuss the mechanisms, similarities and differences, as well as advantages and limitations of different neurotoxin-induced rat models of PD. In the second part of this review, we will discuss the potential future of neurotoxin-induced models of PD. Finally, we will briefly demonstrate the crucial role of gene-environment interactions in PD and discuss fusion or dual PD models. We argue that these models have the potential to significantly further our understanding of PD.
Collapse
Affiliation(s)
- Mohamed El-Gamal
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt. .,Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Mohamed Salama
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Global Brain Health Institute (GBHI), Trinity College Dublin (TCD), Dublin, Ireland
| | | | | | - Ahmed M Fathalla
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Amira M Soliman
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Wael Mohamed
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Mansoura, Egypt.,Department of Basic Medical Science, Kulliyyah of Medicine, International Islamic University, Kuantan, Pahang, Malaysia
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology and Marcs Institute for Brain and Behaviour, Western Sydney University, Sydney, NSW, Australia.,Department of Human Anatomy and Physiology, the Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
38
|
Vedam-Mai V. Harnessing the immune system for the treatment of Parkinson's disease. Brain Res 2021; 1758:147308. [PMID: 33524380 DOI: 10.1016/j.brainres.2021.147308] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/11/2020] [Accepted: 01/16/2021] [Indexed: 01/03/2023]
Abstract
Current treatment options for Parkinson's disease (PD) typically aim to replace dopamine, and hence only provide symptomatic relief. However, in the long run, this approach alone loses its efficacy as it is associated with debilitating side effects. Hence there is an unmet clinical need for addressing levodopa resistant symptoms, and an urgency to develop therapies that can halt or prevent the course of PD. The premise that α-syn can transmit from cell-to-cell in a prion like manner has opened up the possibility for the use of immunotherapy in PD. There is evidence for inflammation in PD as is evidenced by microglial activation, as well as the involvement of the peripheral immune system in PD, and peripheral inflammation can exacerbate dopaminergic degeneration as seen in animal models of the disease. However, mechanisms that link the immune system with PD are not clear, and the sequence of immune responses with respect to PD are still unknown. Nevertheless, our present knowledge offers avenues for the development of immune-based therapies for PD. In order to successfully employ such strategies, we must comprehend the state of the peripheral immune system during the course of PD. This review describes the developments in the field of both active and passive immunotherapies in the treatment of PD, and highlights the crucial need for future research for clarifying the role of inflammation and immunity in this debilitating disease.
Collapse
|
39
|
Liang LP, Fulton R, Bradshaw-Pierce EL, Pearson-Smith J, Day BJ, Patel M. Optimization of Lipophilic Metalloporphyrins Modifies Disease Outcomes in a Rat Model of Parkinsonism. J Pharmacol Exp Ther 2021; 377:1-10. [PMID: 33500265 DOI: 10.1124/jpet.120.000229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/19/2021] [Indexed: 11/22/2022] Open
Abstract
Oxidative stress plays a crucial role in the pathogenesis of Parkinson disease (PD), and one strategy for neuroprotective therapy for PD is to scavenge reactive species using a catalytic antioxidant. Previous studies in our laboratory revealed that pretreatment of lipophilic metalloporphyrins showed protective effects in a mouse PD model. In this study, we optimized the formulations of these metalloporphyrins to deliver them orally and tested their efficacy on disease outcomes in a second species after initiation of an insult (i.e., disease modification). In this study, a pharmaceutical formulation of two metalloporphyrin catalytic antioxidants, AEOL11207 and AEOL11114, was tested for oral drug delivery. Both compounds showed gastrointestinal absorption, achieved high plasma concentrations, and readily penetrated the blood-brain barrier after intravenous or oral delivery. AEOL11207 and AEOL11114 bioavailabilities were calculated to be 24% and 25%, respectively, at a dose of 10 mg/kg via the oral route. In addition, both compounds significantly attenuated 6-hydroxydopamine (6-OHDA)-induced neurotoxic damage, including dopamine depletion, cytokine production, and microglial activation in the striata; dopaminergic neuronal loss in the substantia nigra; oxidative/nitrative stress indices (glutathione disulfide and 3-nitrotyrosine) in the ventral midbrain; and rotation behavioral abnormality in rats. These results indicate that AEOL11207 and AEOL11114 are orally active metalloporphyrins and protect against 6-OHDA neurotoxicity 1-3 days postlesioning, suggesting disease-modifying properties and translational potential for PD. SIGNIFICANCE STATEMENT: Two catalytic antioxidants showed gastrointestinal absorption, achieved high plasma concentrations, and readily penetrated the blood-brain barrier. Both compounds significantly attenuated dopamine depletion, cytokine production, microglial activation, dopaminergic neuronal loss, oxidative/nitrative stress indices, and behavioral abnormality in a Parkinson disease rat model. The results suggest that both metalloporphyrins possess disease-modifying properties that may be useful in treating Parkinson disease.
Collapse
Affiliation(s)
- Li-Ping Liang
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado (L.-P.L., R.F., E.-L.B.-P., J.P.-S., B.J.D., M.P.) and Department of Medicine, National Jewish Health, Denver, Colorado (B.J.D.)
| | - Ruth Fulton
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado (L.-P.L., R.F., E.-L.B.-P., J.P.-S., B.J.D., M.P.) and Department of Medicine, National Jewish Health, Denver, Colorado (B.J.D.)
| | - Erica L Bradshaw-Pierce
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado (L.-P.L., R.F., E.-L.B.-P., J.P.-S., B.J.D., M.P.) and Department of Medicine, National Jewish Health, Denver, Colorado (B.J.D.)
| | - Jennifer Pearson-Smith
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado (L.-P.L., R.F., E.-L.B.-P., J.P.-S., B.J.D., M.P.) and Department of Medicine, National Jewish Health, Denver, Colorado (B.J.D.)
| | - Brian J Day
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado (L.-P.L., R.F., E.-L.B.-P., J.P.-S., B.J.D., M.P.) and Department of Medicine, National Jewish Health, Denver, Colorado (B.J.D.)
| | - Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado (L.-P.L., R.F., E.-L.B.-P., J.P.-S., B.J.D., M.P.) and Department of Medicine, National Jewish Health, Denver, Colorado (B.J.D.)
| |
Collapse
|
40
|
Kline EM, Houser MC, Herrick MK, Seibler P, Klein C, West A, Tansey MG. Genetic and Environmental Factors in Parkinson's Disease Converge on Immune Function and Inflammation. Mov Disord 2021; 36:25-36. [PMID: 33314312 PMCID: PMC8285924 DOI: 10.1002/mds.28411] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/20/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022] Open
Abstract
Idiopathic Parkinson's disease (iPD) is a movement disorder characterized by the degeneration of dopaminergic neurons and aggregation of the protein α-synuclein. Patients with iPD vary in age of symptom onset, rate of progression, severity of motor and non-motor symptoms, and extent of central and peripheral inflammation. Genetic and environmental factors are believed to act synergistically in iPD pathogenesis. We propose that environmental factors (pesticides and infections) increase the risk for iPD via the immune system and that the role of PD risk genes in immune cells is worthy of investigation. This review highlights the major PD-relevant genes expressed in immune cells and key environmental factors that activate immune cells and, alone or in combination with other factors, may contribute to iPD pathogenesis. By reviewing these interactions, we seek to enable the future development of immunomodulatory approaches to prevent or delay onset of iPD. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Elizabeth M Kline
- Laney Graduate School, Emory University, Atlanta, Georgia, USA
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Madelyn C Houser
- Laney Graduate School, Emory University, Atlanta, Georgia, USA
- School of Nursing, Emory University, Atlanta, Georgia, USA
| | - Mary K Herrick
- Laney Graduate School, Emory University, Atlanta, Georgia, USA
- Departments of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Philip Seibler
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Andrew West
- Duke Center for Neurodegeneration and Neurotherapeutics, Duke University, Durham, North Carolina, USA
| | - Malú G Tansey
- Departments of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
41
|
Taguchi D, Ehara A, Kadowaki T, Sakakibara SI, Nakadate K, Hirata K, Ueda S. Minocycline Alleviates Cluster Formation of Activated Microglia and Age-dependent Dopaminergic Cell Death in the Substantia Nigra of Zitter Mutant Rat. Acta Histochem Cytochem 2020; 53:139-146. [PMID: 33437100 PMCID: PMC7785462 DOI: 10.1267/ahc.20-00022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/12/2020] [Indexed: 11/22/2022] Open
Abstract
Microglial activation is a component of neurodegenerative pathology. Here, we examine whether activated microglia participate in age-related dopaminergic (DA) cell death in the substantia nigra pars compacta (SNc) of the zitter (zi/zi) rat, a mutant characterized by deletion of the attractin gene. Confocal microscopy with double-immunohistochemical staining revealed activated microglia-formed cell-clusters surrounding DA neurons in the SNc from 2 weeks after birth. An immunoelectron microscopic study showed that the cytoplasm of activated microglia usually contains phagosome-like vacuoles and lamellar inclusions. Expression levels of the pro-inflammatory cytokines interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS) were increased in the midbrain of 2-month-old zi/zi rats. Chronic treatment with the anti-inflammatory agent minocycline altered the morphology of the microglia, reduced cluster formation by the microglia, and attenuated DA cell death in the SNc, and reduced the expression of IL-1β in the midbrain. These results indicate that activated microglia, at least in part and especially at the initial phase, contribute to DA cell death in the SNc of the zi/zi rat.
Collapse
Affiliation(s)
- Daisuke Taguchi
- Department of Judo Therapy, Faculty of Medical Technology, Teikyo University
- Department of Histology and Neurobiology, Dokkyo Medical University School of Medicine
| | - Ayuka Ehara
- Department of Histology and Neurobiology, Dokkyo Medical University School of Medicine
| | - Taro Kadowaki
- Department of Neurology, Dokkyo Medical University School of Medicine
| | - Shin-ichi Sakakibara
- Laboratory of Molecular Neurobiology, Institute of Applied Brain Sciences, Faculty of Human Sciences, Waseda University
| | - Kazuhiko Nakadate
- Department of Basic Science, Educational and Research Center for Pharmacy, Meiji Pharmaceutical University
| | - Koichi Hirata
- Department of Neurology, Dokkyo Medical University School of Medicine
| | - Shuichi Ueda
- Department of Histology and Neurobiology, Dokkyo Medical University School of Medicine
| |
Collapse
|
42
|
Nehal N, Nabi B, Rehman S, Pathak A, Iqubal A, Khan SA, Yar MS, Parvez S, Baboota S, Ali J. Chitosan coated synergistically engineered nanoemulsion of Ropinirole and nigella oil in the management of Parkinson's disease: Formulation perspective and In vitro and In vivo assessment. Int J Biol Macromol 2020; 167:605-619. [PMID: 33278450 DOI: 10.1016/j.ijbiomac.2020.11.207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/22/2020] [Accepted: 11/29/2020] [Indexed: 12/22/2022]
Abstract
The research presented aims at developing Ropinirole hydrochloride (RHCl) nanoemulsion (NE) with nigella oil for Parkinson's disease (PD). In silico study was done to explore interactions of ropinirole and thymoquinone at receptor site (TNF-α and NFK-β). Ropinirole and Thymoquinone forms a hydrogen bond with residue Arginine 201 and residue Arginine 253 with a bond length of 1.89 Å and 2.30 Å at the NF-κβ receptor. NE was optimized using Central Composite Rotatable Design (CCRD). The globule size of chitosan coated NE, Polydispersity index (PDI) and zeta potential were 183.7 ± 5.2 nm, 0.263 ± 0.005, and 24.9 mV respectively. NE exhibited 85.28% transmittance showing the formulation was clear and transparent. TEM showed that NE had spherical globules with no aggregation. The formulation had a stable pH value of 5.8 ± 0.18. In vitro release and permeation studies exhibited 2 folds and 3.4 folds enhancement when compared with the drug suspension. Neurobehavioral activity and biochemical parameters corroborated well with the pharmacokinetic results. Histopathological study and immunohistochemical analysis were performed to get better picture of 6-OHDA induced toxicity and reversal of PD symptoms. Thus, the NE tailored is a promising synergistic approach yielding enticing outcomes for better management of PD related symptoms.
Collapse
Affiliation(s)
- Nida Nehal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Bushra Nabi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Saleha Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ankita Pathak
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Saif Ahmad Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - M Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
43
|
Kadnikov IA, Verbovaya ER, Voronkov DN, Voronin MV, Seredenin SB. Deferred Administration of Afobazole Induces Sigma1R-Dependent Restoration of Striatal Dopamine Content in a Mouse Model of Parkinson's Disease. Int J Mol Sci 2020; 21:E7620. [PMID: 33076300 PMCID: PMC7593947 DOI: 10.3390/ijms21207620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/28/2020] [Accepted: 10/13/2020] [Indexed: 12/22/2022] Open
Abstract
Previously, we demonstrated that the immediate administration of multitarget anxiolytic afobazole slows down the progression of neuronal damage in a 6-hydroxidodamine (6-OHDA) model of Parkinson's disease due to the activation of chaperone Sigma1R. The aim of the present study is to evaluate the therapeutic potential of deferred afobazole administration in this model. Male ICR mice received a unilateral 6-OHDA lesion of the striatum. Fourteen days after the surgery, mice were treated with afobazole, selective Sigma1R agonist PRE-084, selective Sigma1R antagonist BD-1047, and a combination of BD-1047 with afobazole or PRE-084 for another 14 days. The deferred administration of afobazole restored the intrastriatal dopamine content in the 6-OHDA-lesioned striatum and facilitated motor behavior in rotarod tests. The action of afobazole accorded with the effect of Sigma1R selective agonist PRE-084 and was blocked by Sigma1R selective antagonist BD-1047. The present study illustrates the Sigma1R-dependent effects of afobazole in a 6-OHDA model of Parkinson's disease and reveals the therapeutic potential of Sigma1R agonists in treatment of the condition.
Collapse
Affiliation(s)
- Ilya A. Kadnikov
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya street 8, 125315 Moscow, Russia; (E.R.V.); (M.V.V.)
| | - Ekaterina R. Verbovaya
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya street 8, 125315 Moscow, Russia; (E.R.V.); (M.V.V.)
| | - Dmitry N. Voronkov
- Laboratory of neuromorphology, Research Center of Neurology, Volokolamskoe Highway 80, 125367 Moscow, Russia;
| | - Mikhail V. Voronin
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya street 8, 125315 Moscow, Russia; (E.R.V.); (M.V.V.)
| | - Sergei B. Seredenin
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya street 8, 125315 Moscow, Russia; (E.R.V.); (M.V.V.)
| |
Collapse
|
44
|
Grozdanov V, Danzer KM. Intracellular Alpha-Synuclein and Immune Cell Function. Front Cell Dev Biol 2020; 8:562692. [PMID: 33178682 PMCID: PMC7594520 DOI: 10.3389/fcell.2020.562692] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/24/2020] [Indexed: 11/13/2022] Open
Abstract
Intracellular alpha-synuclein has numerous effects on different functions of the cell. Although it is expressed in a wide spectrum of cell types from different lineages, most of our knowledge about it was generated by studying neuronal or glial cells. However, the role of immune cells in Parkinson's disease and related synucleinopathies has recently emerged. Altered immune cell phenotypes and functions have been reported not only in animal models, but also in human disease. While the response of immune cells to extracellular alpha-synuclein has been thoroughly studied, insights into the effects of endogenously expressed or taken-up alpha-synuclein on the function of immune cells remain scarce. Such insights may prove to be important for understanding the complex cellular and molecular events resulting in neurodegeneration and aid the development of novel therapies. We review the current state of knowledge about how alpha-synuclein and its pathologic manifestations affect the phenotype and function of peripheral and central nervous system (CNS) immune cells, and discuss the potential of this topic for advancing our understanding of synucleinopathies.
Collapse
|
45
|
Airavaara M, Parkkinen I, Konovalova J, Albert K, Chmielarz P, Domanskyi A. Back and to the Future: From Neurotoxin-Induced to Human Parkinson's Disease Models. ACTA ACUST UNITED AC 2020; 91:e88. [PMID: 32049438 DOI: 10.1002/cpns.88] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disorder characterized by motor symptoms such as tremor, slowness of movement, rigidity, and postural instability, as well as non-motor features like sleep disturbances, loss of ability to smell, depression, constipation, and pain. Motor symptoms are caused by depletion of dopamine in the striatum due to the progressive loss of dopamine neurons in the substantia nigra pars compacta. Approximately 10% of PD cases are familial arising from genetic mutations in α-synuclein, LRRK2, DJ-1, PINK1, parkin, and several other proteins. The majority of PD cases are, however, idiopathic, i.e., having no clear etiology. PD is characterized by progressive accumulation of insoluble inclusions, known as Lewy bodies, mostly composed of α-synuclein and membrane components. The cause of PD is currently attributed to cellular proteostasis deregulation and mitochondrial dysfunction, which are likely interdependent. In addition, neuroinflammation is present in brains of PD patients, but whether it is the cause or consequence of neurodegeneration remains to be studied. Rodents do not develop PD or PD-like motor symptoms spontaneously; however, neurotoxins, genetic mutations, viral vector-mediated transgene expression and, recently, injections of misfolded α-synuclein have been successfully utilized to model certain aspects of the disease. Here, we critically review the advantages and drawbacks of rodent PD models and discuss approaches to advance pre-clinical PD research towards successful disease-modifying therapy. © 2020 The Authors.
Collapse
Affiliation(s)
- Mikko Airavaara
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ilmari Parkkinen
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Julia Konovalova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Katrina Albert
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
46
|
Lee MY, Lin BF, Chan MH, Chen HH. Increased behavioral and neuronal responses to a hallucinogenic drug after adolescent toluene exposure in mice: Effects of antipsychotic treatment. Toxicology 2020; 445:152602. [PMID: 32980479 DOI: 10.1016/j.tox.2020.152602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/19/2020] [Accepted: 09/22/2020] [Indexed: 01/23/2023]
Abstract
Toluene has been characterized as a non-classical hallucinogen drug through activation of 5-HT2A receptors and antagonism of NMDA receptors. It remains unclear whether psychotic symptoms after long-term and intense toluene exposure are associated with abnormalities in 5-HT2A receptor function. The present study examined whether the responses to a hallucinogenic 5-HT2A receptor agonist 2,5-dimethoxy-4-iodoamphetamine (DOI) were altered in a mouse model of toluene psychosis. Male NMRI mice were subchronically treated with toluene during adolescence. Reciprocal social interaction test and novel object recognition test were conducted to confirm the persistent behavioral deficits in adulthood. Subsequently, DOI-induced head twitch, c-Fos and Egr-2 expression, field potentials in the medial prefrontal cortex (mPFC), and the levels of 5-HT2A, 5-HT1A and mGlu2 receptors in the mPFC were monitored. Toluene exposure during adolescence produced social and memory impairments and enhanced DOI-induced behavioral, molecular and electrophysiological responses, but did not change the levels of 5-HT2A, 5-HT1A or mGlu2 receptors in the mPFC. Moreover, the effects of haloperidol and risperidone on the behavioral deficits and hyper-responsiveness to DOI after adolescent toluene exposure were compared. When administered after adolescent toluene exposure, risperidone could reverse social withdrawal, cognitive impairment and hypersensitivity to DOI, whereas haloperidol was only beneficial for social withdrawal. These findings suggest that increased functionality of 5-HT2A receptors may play a critical role in solvent-induced psychosis and recommend the antipsychotics with more selective 5-HT2A receptor antagonism as the first-line treatment for solvent-induced psychosis.
Collapse
Affiliation(s)
- Mei-Yi Lee
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Rd. Zhunan, Miaoli, 35053, Taiwan
| | - Bih-Fen Lin
- Department of Laboratory Medicine and Biotechnology School of Medicine, Tzu Chi University, No. 701, Sec. 3, Zhongyang Rd., Hualien, 97004, Taiwan
| | - Ming-Huan Chan
- Institute of Neuroscience, National Chengchi University, NO. 64, Sec. 2, Zhinan Rd., Taipei, 11605, Taiwan; Research Center for Mind, Brain, and Learning, National Chengchi University, NO. 64, Sec. 2, Zhinan Rd., Taipei, 11605, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan.
| | - Hwei-Hsien Chen
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Rd. Zhunan, Miaoli, 35053, Taiwan; Institute of Neuroscience, National Chengchi University, NO. 64, Sec. 2, Zhinan Rd., Taipei, 11605, Taiwan; Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, No. 145, Xingda Rd., Taichung, 40227, Taiwan.
| |
Collapse
|
47
|
Haque A, Samantaray S, Knaryan VH, Capone M, Hossain A, Matzelle D, Chandran R, Shields DC, Farrand AQ, Boger HA, Banik NL. Calpain mediated expansion of CD4+ cytotoxic T cells in rodent models of Parkinson's disease. Exp Neurol 2020; 330:113315. [PMID: 32302678 PMCID: PMC7282933 DOI: 10.1016/j.expneurol.2020.113315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/17/2020] [Accepted: 04/13/2020] [Indexed: 01/11/2023]
Abstract
Parkinson's disease (PD), a debilitating progressive degenerative movement disorder associated with loss of dopaminergic (DA) neurons in the substantia nigra (SN), afflicts approximately one million people in the U.S., including a significant number of Veterans. Disease characteristics include tremor, rigidity, postural instability, bradykinesia, and at a cellular level, glial cell activation and Lewy body inclusions in DA neurons. The most potent medical/surgical treatments do not ultimately prevent disease progression. Therefore, new therapies must be developed to halt progression of the disease. While the mechanisms of the degenerative process in PD remain elusive, chronic inflammation, a common factor in many neurodegenerative diseases, has been implicated with associated accumulation of toxic aggregated α-synuclein in neurons. Calpain, a calcium-activated cysteine neutral protease, plays a pivotal role in SN and spinal cord degeneration in PD via its role in α-synuclein aggregation, activation/migration of microglia and T cells, and upregulation of inflammatory processes. Here we report an increased expression of a subset of CD4+ T cells in rodent models of PD, including MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mice and DSP-4 [N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride]/6-hydroxydopamine rats, which produced higher levels of perforin and granzyme B - typically found in cytotoxic T cells. Importantly, the CD4+ cytotoxic subtype was attenuated following calpain inhibition in MPTP mice, suggesting that calpain and this distinct CD4+ T cell subset may have critical roles in the inflammatory process, disease progression, and neurodegeneration in PD.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Supriti Samantaray
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA
| | - Varduhi H Knaryan
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA
| | - Mollie Capone
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Azim Hossain
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC 29401, USA
| | - Raghavendar Chandran
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA
| | - Donald C Shields
- Department of Neurosurgery, The George Washington University, Washington, DC, USA
| | - Ariana Q Farrand
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Heather A Boger
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC 29401, USA.
| |
Collapse
|
48
|
San Luciano M, Tanner CM, Meng C, Marras C, Goldman SM, Lang AE, Tolosa E, Schüle B, Langston JW, Brice A, Corvol JC, Goldwurm S, Klein C, Brockman S, Berg D, Brockmann K, Ferreira JJ, Tazir M, Mellick GD, Sue CM, Hasegawa K, Tan EK, Bressman S, Saunders-Pullman R. Nonsteroidal Anti-inflammatory Use and LRRK2 Parkinson's Disease Penetrance. Mov Disord 2020; 35:1755-1764. [PMID: 32662532 DOI: 10.1002/mds.28189] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/15/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The penetrance of leucine rich repeat kinase 2 (LRRK2) mutations is incomplete and may be influenced by environmental and/or other genetic factors. Nonsteroidal anti-inflammatory drugs (NSAIDs) are known to reduce inflammation and may lower Parkinson's disease (PD) risk, but their role in LRRK2-associated PD is unknown. OBJECTIVES The objective of this study is to evaluate the association of regular NSAID use and LRRK2-associated PD. METHODS Symptomatic ("LRRK2-PD") and asymptomatic ("LRRK2-non-PD") participants with LRRK2 G2019S, R1441X, or I2020T variants (definitely pathogenic variant carriers) or G2385R or R1628P variants (risk variant carriers) from 2 international cohorts provided information on regular ibuprofen and/or aspirin use (≥2 pills/week for ≥6 months) prior to the index date (diagnosis date for PD, interview date for non-PD). Multivariate logistic regression was used to evaluate the relationship between regular NSAID use and PD for any NSAID, separately for ibuprofen and aspirin in all carriers and separately in pathogenic and risk variant groups. RESULTS A total of 259 LRRK2-PD and 318 LRRK2-non-PD participants were enrolled. Regular NSAID use was associated with reduced odds of PD in the overall cohort (odds ratio [OR], 0.34; 95% confidence interval [CI], 0.21-0.57) and in both pathogenic and risk variant carriers (ORPathogenic , 0.38; 95% CI, 0.21-0.67 and ORRiskVariant , 0.19; 95% CI, 0.04-0.99). Similar associations were observed for ibuprofen and aspirin separately (ORIbuprofen , 0.19; 95% CI, 0.07-0.50 and ORAspirin , 0.51; 95% CI, 0.28-0.91). CONCLUSIONS Regular NSAID use may be associated with reduced penetrance in LRRK2-associated PD. The LRRK2 protein is involved in inflammatory pathways and appears to be modulated by regular anti-inflammatory use. Longitudinal observational and interventional studies of NSAID exposure and LRRK2-PD are needed to confirm this association. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Marta San Luciano
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Caroline M Tanner
- Department of Neurology, University of California San Francisco, San Francisco, California, USA.,Department of Neurology, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Cheryl Meng
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Connie Marras
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Centre, Toronto, Ontario, Canada.,Department of Neurology, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Samuel M Goldman
- Department of Neurology, University of California San Francisco, San Francisco, California, USA.,Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Anthony E Lang
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Centre, Toronto, Ontario, Canada.,Department of Neurology, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Eduardo Tolosa
- Movement Disorders Unit, Neurology Service, Hospital Clínic, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (IDIBAPS) Universitat de Barcelona, Catalonia, Spain
| | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - J William Langston
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA.,Department of Neurology, Stanford University School of Medicine, Stanford, California, USA
| | - Alexis Brice
- Sorbonne Universites, UPMC Universite Paris 6 UMR_S 1127, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle epiniere, ICM, Paris, France
| | - Jean-Christophe Corvol
- Sorbonne Universites, UPMC Universite Paris 6 UMR_S 1127, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle epiniere, ICM, Paris, France
| | | | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Simone Brockman
- School of Psychiatry and Clinical Neurosciences, University of Western Australia and Fremantle Hospital, Western Australia, Australia
| | - Daniela Berg
- Department for Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Kathrin Brockmann
- Department for Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Joachim J Ferreira
- Clinical Pharmacology Unit, Instituto de Medicina Molecular, Lisbon, Portugal
| | - Meriem Tazir
- Service de Neurologie CHU Mustapha, Alger, Algeria
| | - George D Mellick
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Australia
| | - Carolyn M Sue
- Department of Neurogenetics, Kolling Institute, University of Sydney, Sydney, Australia
| | - Kazuko Hasegawa
- Department of Neurology, Sagamihara National Hospital, Kanagawa, Japan
| | - Eng King Tan
- Department of Neurology, Singapore General Hospital, Singapore
| | - Susan Bressman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | |
Collapse
|
49
|
Belloli S, Morari M, Murtaj V, Valtorta S, Moresco RM, Gilardi MC. Translation Imaging in Parkinson's Disease: Focus on Neuroinflammation. Front Aging Neurosci 2020; 12:152. [PMID: 32581765 PMCID: PMC7289967 DOI: 10.3389/fnagi.2020.00152] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the appearance of α-synuclein insoluble aggregates known as Lewy bodies. Neurodegeneration is accompanied by neuroinflammation mediated by cytokines and chemokines produced by the activated microglia. Several studies demonstrated that such an inflammatory process is an early event, and contributes to oxidative stress and mitochondrial dysfunctions. α-synuclein fibrillization and aggregation activate microglia and contribute to disease onset and progression. Mutations in different genes exacerbate the inflammatory phenotype in the monogenic compared to sporadic forms of PD. Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) with selected radiopharmaceuticals allow in vivo imaging of molecular modifications in the brain of living subjects. Several publications showed a reduction of dopaminergic terminals and dopamine (DA) content in the basal ganglia, starting from the early stages of the disease. Moreover, non-dopaminergic neuronal pathways are also affected, as shown by in vivo studies with serotonergic and glutamatergic radiotracers. The role played by the immune system during illness progression could be investigated with PET ligands that target the microglia/macrophage Translocator protein (TSPO) receptor. These agents have been used in PD patients and rodent models, although often without attempting correlations with other molecular or functional parameters. For example, neurodegeneration and brain plasticity can be monitored using the metabolic marker 2-Deoxy-2-[18F]fluoroglucose ([18F]-FDG), while oxidative stress can be probed using the copper-labeled diacetyl-bis(N-methyl-thiosemicarbazone) ([Cu]-ATSM) radioligand, whose striatal-specific binding ratio in PD patients seems to correlate with a disease rating scale and motor scores. Also, structural and functional modifications during disease progression may be evaluated by Magnetic Resonance Imaging (MRI), using different parameters as iron content or cerebral volume. In this review article, we propose an overview of in vivo clinical and non-clinical imaging research on neuroinflammation as an emerging marker of early PD. We also discuss how multimodal-imaging approaches could provide more insights into the role of the inflammatory process and related events in PD development.
Collapse
Affiliation(s)
- Sara Belloli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Michele Morari
- Section of Pharmacology, Department of Medical Sciences, National Institute for Neuroscience, University of Ferrara, Ferrara, Italy
| | - Valentina Murtaj
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Silvia Valtorta
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Rosa Maria Moresco
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Maria Carla Gilardi
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
50
|
Elfil M, Kamel S, Kandil M, Koo BB, Schaefer SM. Implications of the Gut Microbiome in Parkinson's Disease. Mov Disord 2020; 35:921-933. [DOI: 10.1002/mds.28004] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/06/2020] [Accepted: 02/04/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Mohamed Elfil
- Department of NeurologyYale University New Haven Connecticut USA
| | - Serageldin Kamel
- Department of NeurologyYale University New Haven Connecticut USA
| | - Mohamed Kandil
- Department of NeurologyYale University New Haven Connecticut USA
| | - Brian B. Koo
- Department of NeurologyYale University New Haven Connecticut USA
- Center for Neuroepidemiology and Clinical Neurologic Research Yale New Haven Connecticut USA
- Department of NeurologyConnecticut Veterans Affairs Healthcare System West Haven Connecticut USA
| | - Sara M. Schaefer
- Department of NeurologyYale University New Haven Connecticut USA
| |
Collapse
|