1
|
Negro-Demontel L, Maleki AF, Reich DS, Kemper C. The complement system in neurodegenerative and inflammatory diseases of the central nervous system. Front Neurol 2024; 15:1396520. [PMID: 39022733 PMCID: PMC11252048 DOI: 10.3389/fneur.2024.1396520] [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: 03/05/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Neurodegenerative and neuroinflammatory diseases, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis, affect millions of people globally. As aging is a major risk factor for neurodegenerative diseases, the continuous increase in the elderly population across Western societies is also associated with a rising prevalence of these debilitating conditions. The complement system, a crucial component of the innate immune response, has gained increasing attention for its multifaceted involvement in the normal development of the central nervous system (CNS) and the brain but also as a pathogenic driver in several neuroinflammatory disease states. Although complement is generally understood as a liver-derived and blood or interstitial fluid operative system protecting against bloodborne pathogens or threats, recent research, particularly on the role of complement in the healthy and diseased CNS, has demonstrated the importance of locally produced and activated complement components. Here, we provide a succinct overview over the known beneficial and pathological roles of complement in the CNS with focus on local sources of complement, including a discussion on the potential importance of the recently discovered intracellularly active complement system for CNS biology and on infection-triggered neurodegeneration.
Collapse
Affiliation(s)
- Luciana Negro-Demontel
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, United States
- Department of Histology and Embryology, Faculty of Medicine, UDELAR, Montevideo, Uruguay
- Neuroinflammation and Gene Therapy Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Adam F. Maleki
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, United States
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD, United States
| | - Daniel S. Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD, United States
| | - Claudia Kemper
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, United States
| |
Collapse
|
2
|
Yuan X, Wan L, Chen Z, Long Z, Chen D, Liu P, Fu Y, Zhu S, Peng L, Qiu R, Tang B, Jiang H. Peripheral Inflammatory and Immune Landscape in Multiple System Atrophy: A Cross-Sectional Study. Mov Disord 2024; 39:391-399. [PMID: 38155513 DOI: 10.1002/mds.29674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/16/2023] [Accepted: 11/09/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND Neuroinflammation might contribute to the pathogenesis of multiple systemic atrophy (MSA). However, specific alterations in the peripheral inflammatory and immune profiles of patients with MSA remain unclear. OBJECTIVES To determine the peripheral inflammatory and immune profiles of patients with MSA and their potential value as biomarkers for facilitating clinical diagnosis and monitoring disease severity. METHODS This cross-sectional study included 235, 240, and 235 patients with MSA, patients with Parkinson's disease (PD), and healthy controls (HCs), respectively. Inflammatory and immune parameters were measured in peripheral blood, differences between groups were assessed, and clusters were analyzed. Associations between the parameters and clinical characteristics of MSA were assessed using Spearman and partial correlation analyses. RESULTS Significant differences were observed especially in monocytes, neutrophils-to-lymphocyte ratio (NLR) and neutrophils-to-lymphocyte ratio (MPV) between MSA patients and HCs (P < 0.01). Monocytes and uric acid (UA) levels were also significantly different between the MSA and PD patients (P < 0.05). The combination of NLR and MPV distinguished MSA-P patients from HCs (areas under the curve = 0.824). In addition, complement components C4 and C3 were significantly correlated with the Scale Outcomes in PD for Autonomic Symptoms and Wexner scale, whereas immunoglobulin G (IgG) was significantly correlated with scores of Unified Multiple System Atrophy Rating Scale (P < 0.05). CONCLUSIONS In MSA patients, monocytes, NLR and MPV might serve as potential diagnostic biomarkers, whereas MLR, C3, C4, and IgG significantly correlate with disease severity. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xinrong Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Zhe Long
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Daji Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Panyan Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - You Fu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Sudan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Linliu Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
3
|
Contaldi E, Magistrelli L, Comi C. Disease mechanisms as subtypes: Immune dysfunction in Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:67-93. [PMID: 36803824 DOI: 10.1016/b978-0-323-85555-6.00008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
In recent years, the contraposition between inflammatory and neurodegenerative processes has been increasingly challenged. Inflammation has been emphasized as a key player in the onset and progression of Parkinson disease (PD) and other neurodegenerative disorders. The strongest indicators of the involvement of the immune system derived from evidence of microglial activation, profound imbalance in phenotype and composition of peripheral immune cells, and impaired humoral immune responses. Moreover, peripheral inflammatory mechanisms (e.g., involving the gut-brain axis) and immunogenetic factors are likely to be implicated. Even though several lines of preclinical and clinical studies are supporting and defining the complex relationship between the immune system and PD, the exact mechanisms are currently unknown. Similarly, the temporal and causal connections between innate and adaptive immune responses and neurodegeneration are unsettled, challenging our ambition to define an integrated and holistic model of the disease. Despite these difficulties, current evidence is providing the unique opportunity to develop immune-targeted approaches for PD, thus enriching our therapeutic armamentarium. This chapter aims to provide an extensive overview of past and present studies that explored the implication of the immune system in neurodegeneration, thus paving the road for the concept of disease modification in PD.
Collapse
Affiliation(s)
- Elena Contaldi
- Movement Disorders Centre, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Luca Magistrelli
- Movement Disorders Centre, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Cristoforo Comi
- Neurology Unit, S.Andrea Hospital, Department of Translational Medicine, University of Piemonte Orientale, Vercelli, Italy.
| |
Collapse
|
4
|
Sušjan-Leite P, Ramuta TŽ, Boršić E, Orehek S, Hafner-Bratkovič I. Supramolecular organizing centers at the interface of inflammation and neurodegeneration. Front Immunol 2022; 13:940969. [PMID: 35979366 PMCID: PMC9377691 DOI: 10.3389/fimmu.2022.940969] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
The pathogenesis of neurodegenerative diseases involves the accumulation of misfolded protein aggregates. These deposits are both directly toxic to neurons, invoking loss of cell connectivity and cell death, and recognized by innate sensors that upon activation release neurotoxic cytokines, chemokines, and various reactive species. This neuroinflammation is propagated through signaling cascades where activated sensors/receptors, adaptors, and effectors associate into multiprotein complexes known as supramolecular organizing centers (SMOCs). This review provides a comprehensive overview of the SMOCs, involved in neuroinflammation and neurotoxicity, such as myddosomes, inflammasomes, and necrosomes, their assembly, and evidence for their involvement in common neurodegenerative diseases. We discuss the multifaceted role of neuroinflammation in the progression of neurodegeneration. Recent progress in the understanding of particular SMOC participation in common neurodegenerative diseases such as Alzheimer's disease offers novel therapeutic strategies for currently absent disease-modifying treatments.
Collapse
Affiliation(s)
- Petra Sušjan-Leite
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Taja Železnik Ramuta
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Elvira Boršić
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Sara Orehek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Iva Hafner-Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Ljubljana, Slovenia
| |
Collapse
|
5
|
Ganguly U, Singh S, Chakrabarti S, Saini AK, Saini RV. Immunotherapeutic interventions in Parkinson's disease: Focus on α-Synuclein. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 129:381-433. [PMID: 35305723 DOI: 10.1016/bs.apcsb.2021.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized classically by motor manifestations. However, nonmotor symptoms appear early in the course of the disease progression, making both diagnosis and treatment difficult. The pathology of PD is complicated by the accumulation and aggregation of misfolded proteins in intracellular cytoplasmic inclusions called Lewy bodies (LBs). The main toxic component of LBs is the protein α-Synuclein which plays a pivotal role in PD pathogenesis. α-Synuclein can propagate from cell-to-cell exhibiting prion-like properties and spread PD pathology throughout the central nervous system. Immunotherapeutic interventions in PD, both active and passive immunization, have targeted α-Synuclein in both experimental models and clinical trials. In addition, targeting the hyperactive inflammation in PD also holds promise in designing potential immunotherapeutics. The inflammatory and proteotoxic pathways are interlinked and contribute immensely to the disease pathology. In this chapter, we critically review the targets of immunotherapeutic interventions in PD, focusing on the pathogenetic mechanisms of PD, particularly neuroinflammation and α-Synuclein misfolding, aggregation, and propagation. We thoroughly summarized the various immunotherapeutic strategies designed to treat PD-in vitro, in vivo, and clinical trials. The development of these targeted immunotherapies could open a new avenue in the treatment of patients with PD.
Collapse
Affiliation(s)
- Upasana Ganguly
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, India
| | - Sukhpal Singh
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, India
| | - Sasanka Chakrabarti
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, India
| | - Adesh K Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, India
| | - Reena V Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, India.
| |
Collapse
|
6
|
Ziabska K, Ziemka-Nalecz M, Pawelec P, Sypecka J, Zalewska T. Aberrant Complement System Activation in Neurological Disorders. Int J Mol Sci 2021; 22:4675. [PMID: 33925147 PMCID: PMC8125564 DOI: 10.3390/ijms22094675] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
The complement system is an assembly of proteins that collectively participate in the functions of the healthy and diseased brain. The complement system plays an important role in the maintenance of uninjured (healthy) brain homeostasis, contributing to the clearance of invading pathogens and apoptotic cells, and limiting the inflammatory immune response. However, overactivation or underregulation of the entire complement cascade within the brain may lead to neuronal damage and disturbances in brain function. During the last decade, there has been a growing interest in the role that this cascading pathway plays in the neuropathology of a diverse array of brain disorders (e.g., acute neurotraumatic insult, chronic neurodegenerative diseases, and psychiatric disturbances) in which interruption of neuronal homeostasis triggers complement activation. Dysfunction of the complement promotes a disease-specific response that may have either beneficial or detrimental effects. Despite recent advances, the explicit link between complement component regulation and brain disorders remains unclear. Therefore, a comprehensible understanding of such relationships at different stages of diseases could provide new insight into potential therapeutic targets to ameliorate or slow progression of currently intractable disorders in the nervous system. Hence, the aim of this review is to provide a summary of the literature on the emerging role of the complement system in certain brain disorders.
Collapse
Affiliation(s)
| | | | | | | | - Teresa Zalewska
- Mossakowski Medical Research Centre, NeuroRepair Department, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland; (K.Z.); (M.Z.-N.); (P.P.); (J.S.)
| |
Collapse
|
7
|
Role of microgliosis, oxidative stress and associated neuroinflammation in the pathogenesis of Parkinson's disease: The therapeutic role of Nrf2 activators. Neurochem Int 2021; 145:105014. [PMID: 33689805 DOI: 10.1016/j.neuint.2021.105014] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 12/24/2022]
Abstract
Microglial cells are the resident immune cells of the central nervous system. They are essential for normal functioning, maintenance of tissue integrity, clearance of dying neurons, elimination of pathogens, development and maintenance of homeostasis of the CNS. Many studies have consistently reported that oxidative stress and associated neuroinflammation mediated by microglial cells have a degenerating effect on dopaminergic neurons. In Parkinson's disease, the microglial cells by a process called microgliosis undergo rapid proliferation, accumulate at the site of tissue injury and undergo phenotypic and functional changes that result in the release of massive amounts of free radicals causing inflammation and neurodegeneration of dopaminergic neurons. Following the discovery of the irrefutable role oxidative stress and associated neuroinflammation, several proven antioxidants were tested for possible protective and therapeutic potential in Parkinson's disease but the results so far have not been encouraging and equivocal. Consequently, it is rational to look for endogenous targets that enhance the oxidative defense mechanism against free radicals and protect dopaminergic neurons from neuroinflammation and neurodegeneration. One such target is a nuclear factor-erythroid -2-related factor 2 (Nrf2). Nrf2 is a redox-sensitive transcription factor located in the cytoplasm of the cells that helps cells adapt to oxidative stress and inflammation by upregulating the expression of almost 200 cytoprotective genes. Fractalkine exists in a transmembrane form and a soluble form and is a cytokine that links microglial cells and Nrf2. The fractalkine receptors, expressed exclusively by microglial cells, on activation by fractalkine protects dopaminergic neurons from degeneration caused by free radicals and pro-inflammatory mediators through increased expression of Nrf2 dependent genes. The current anti Parkinsonism drugs do not cure the disease and also cause several debilitating motor and non-motor adverse drug effects. So it becomes imperative to explore novel targets and discover novel therapeutic agents to treat Parkinson's disease in a better way and improve the quality of life of patients with Parkinson's disease.
Collapse
|
8
|
Cheng J, Liao Y, Dong Y, Hu H, Yang N, Kong X, Li S, Li X, Guo J, Qin L, Yu J, Ma C, Li J, Li M, Tang B, Yuan Z. Microglial autophagy defect causes parkinson disease-like symptoms by accelerating inflammasome activation in mice. Autophagy 2020; 16:2193-2205. [PMID: 32003282 PMCID: PMC7751565 DOI: 10.1080/15548627.2020.1719723] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 10/17/2019] [Accepted: 01/17/2020] [Indexed: 02/07/2023] Open
Abstract
Microglial activation-induced neuroinflammation is closely associated with the development of Parkinson disease (PD). Macroautophagy/autophagy regulates many biological processes, but the role of autophagy in microglial activation during PD development remains largely unclear. In this study, we showed that deletion of microglial Atg5 caused PD-like symptoms in mice, characterized by impairment in motor coordination and cognitive learning, loss of tyrosine hydroxylase (TH) neurons, enhancement of neuroinflammation and reduction in dopamine levels in the striatum. Mechanistically, we found that inhibition of autophagy led to NLRP3 (NLR family pyrin domain containing 3) inflammasome activation via PDE10A (phosphodiesterase 10A)-cyclic adenosine monophosphate (cAMP) signaling in microglia, and the sequential upregulation of downstream IL1B/IL-1β in turn increased the expression of MIF (macrophage migration inhibitory factor [glycosylation-inhibiting factor]), a pro-inflammatory cytokine. Inhibition of NLRP3 inflammasome activation by administration of MCC950, a specific inhibitor for NLRP3, decreased MIF expression and neuroinflammatory levels, and rescued the loss of TH neurons in the substantial nigra (SN). Interestingly, we found that serum MIF levels in PD patients were significantly elevated. Taken together, our results reveal an important role of autophagy in microglial activation-driven PD-like symptoms, thus providing potential targets for the clinical treatment of PD. Abbreviations: ATG: autophagy related; cAMP: cyclic adenosine monophosphate; cKO: conditional knockout; NOS2/INOS: nitric oxide synthase 2, inducible; IL1B: interleukin 1 beta; ITGAM/CD-11b: integrin alpha M/cluster of differentiation molecule 11B; MAP1LC3: microtubule-associated protein 1 light chain 3; MIF: macrophage migration inhibitory factor (glycosylation-inhibiting factor); NLRP3: NLR family pyrin domain containing 3; PBS: phosphate-buffered saline; PD: parkinson disease; PDE10A: phosphodiesterase 10A; SN: substantial nigra; TH: tyrosine hydroxylase; TNF: tumor necrosis factor; WT: wild type.
Collapse
Affiliation(s)
- Jinbo Cheng
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, China
| | - Yajin Liao
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, China
| | - Yuan Dong
- Department of Biochemistry, Medical College, Qingdao University, Qingdao, Shandong, China
| | - Han Hu
- Institute of Apicultural Research, Chinese Academy of Agricultural Science, Beijing, China
| | - Nannan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangxi Kong
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Shuoshuo Li
- The State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaoheng Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorder, Central South University, Changsha, Hunan, China
| | - Lixia Qin
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiezhong Yu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Cungen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jianke Li
- Institute of Apicultural Research, Chinese Academy of Agricultural Science, Beijing, China
| | - Mingtao Li
- Department of Pharmacology and the Proteomics Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorder, Central South University, Changsha, Hunan, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
- Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
9
|
Zhou Y, Su Y, Xu W, Wang W, Yao S. Constipation Increases Disability and Decreases Dopamine Levels in the Nigrostriatal System through Gastric Inflammatory Factors in Parkinson's Disease. Curr Neurovasc Res 2020; 16:241-249. [PMID: 31258082 DOI: 10.2174/1567202616666190618170103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Recent studies suggest that not only is constipation a clinical marker of premotor phase in Parkinson's Disease (PD), but is also correlated with the duration and severity. Some reports indicated that inflammatory from gut dysbiosis might be involved in the pathogenesis of PD, but the correlation between them remains poorly understood. This study aims to investigate how the presence of constipation affects the dopamine level of nigrostriatal system and whether gastrointestinal (GI) inflammation is involved in the brain-gut axis. METHODS Clinical materials, serum inflammatory factors, and datum of dopamine level including 84 cases and 83 controls, were collected consecutively and randomly from November 1, 2017 to October 31, 2018. Dopamine levels of nigrostriatal system were detected by [18F]-DTBZ radiotracer (18F-AV-133). Data analysis was conducted by variance, covariance analysis, bicorrelation, partial correlation, chi-square analysis and logistic regression. RESULTS The mean age of cases was older than that of controls, and male predominance was also observed (P<0.05). The mean scores of Hoehn-Yahr and unified Parkinson's disease rating scale Ⅲ (UPDRS-Ⅲ) were of significantly different duration between two groups (P<0.05). The total dose of levodopa was not different between two groups (P>0.05). The dopamine levels of putamen and caudate nucleus, especially in the dorsal part of putamen, were significantly decreased in cases than that in controls (P<0.05). There were significant differences of complement 3 (C3) and complement 4 (C4) between cases and controls (P<0.05). Dopamine levels in putamen and caudate nucleus were negatively correlated with serum concentrations of immunoglobulin A (IgA), immunoglobulin G (IgG) and C3 in cases (P<0.05). But we did not observe similar negative correlations in controls (P>0.05). CONCLUSION The presence of constipation may increase the severity of motor symptoms and decrease dopamine levels of nigrostriatal system in PD. Inflammatory factors may be involved in the brain-gut axis of PD.
Collapse
Affiliation(s)
- Yongtao Zhou
- The Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China.,The Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China.,Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing 100029, China
| | - Yusheng Su
- The Nuclear Medicine Department, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Weihua Xu
- Gastroenterology Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Beijing 100053, China
| | - Wei Wang
- Gastroenterology Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Beijing 100053, China
| | - Shukun Yao
- The Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China.,Gastroenterology Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Beijing 100053, China
| |
Collapse
|
10
|
Sim KY, Im KC, Park SG. The Functional Roles and Applications of Immunoglobulins in Neurodegenerative Disease. Int J Mol Sci 2020; 21:E5295. [PMID: 32722559 PMCID: PMC7432158 DOI: 10.3390/ijms21155295] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Natural autoantibodies, immunoglobulins (Igs) that target self-proteins, are common in the plasma of healthy individuals; some of the autoantibodies play pathogenic roles in systemic or tissue-specific autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus. Recently, the field of autoantibody-associated diseases has expanded to encompass neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD), with related studies examining the functions of Igs in the central nervous system (CNS). Recent evidence suggests that Igs have various effects in the CNS; these effects are associated with the prevention of neurodegeneration, as well as induction. Here, we summarize the functional roles of Igs with respect to neurodegenerative disease (AD and PD), focusing on the target antigens and effector cell types. In addition, we review the current knowledge about the roles of these antibodies as diagnostic markers and immunotherapies.
Collapse
Affiliation(s)
| | | | - Sung-Gyoo Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea; (K.-Y.S.); (K.C.I.)
| |
Collapse
|
11
|
Ding X, Yang W, Ren Q, Hu J, Yang S, Han W, Wang J, Wang X, Wang H. Serum IgG-induced microglial activation enhances neuronal cytolysis via the NO/sGC/PKG pathway in children with opsoclonus-myoclonus syndrome and neuroblastoma. J Neuroinflammation 2020; 17:190. [PMID: 32546235 PMCID: PMC7298801 DOI: 10.1186/s12974-020-01839-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 05/11/2020] [Indexed: 12/14/2022] Open
Abstract
Background Opsoclonus-myoclonus syndrome (OMS) is a rare neurological disease. Some children with OMS also have neuroblastoma (NB). We and others have previously documented that serum IgG from children with OMS and NB induces neuronal cytolysis and activates several signaling pathways. However, the mechanisms underlying OMS remain unclear. Here, we investigated whether nitric oxide (NO) from activated microglias and its cascade contribute to neuronal cytolysis in pediatric OMS. Methods The activation of cultured cerebral cortical and cerebellar microglias incubated with sera or IgG isolated from sera of children with OMS and NB was measured by the expression of the activation marker, cytokines, and NO. Neuronal cytolysis was determined after exposing to IgG-treated microglia-conditioned media. Using inhibitors and activators, the effects of NO synthesis and its intracellular cascade, namely soluble guanylyl cyclase (sGC) and protein kinase G (PKG), on neuronal cytolysis were evaluated. Results Incubation with sera or IgG from children with OMS and NB increased the activation of cerebral cortical and cerebellar microglias, but not the activation of astrocytes or the cytolysis of glial cells. Moreover, the cytolysis of neurons was elevated by conditioned media from microglias incubated with IgG from children with OMS and NB. Furthermore, the expression of NO, sGC, and PKG was increased. Neuronal cytolysis was relieved by the inhibitors of NO signaling, while neuronal cytolysis was exacerbated by the activators of NO signaling but not proinflammatory cytokines. The cytolysis of neurons was suppressed by pretreatment with the microglial inhibitor minocycline, a clinically tested drug. Finally, increased microglial activation did not depend on the Fab fragment of serum IgG. Conclusions Serum IgG from children with OMS and NB potentiates microglial activation, which induces neuronal cytolysis through the NO/sGC/PKG pathway, suggesting an applicability of microglial inhibitor as a therapeutic candidate.
Collapse
Affiliation(s)
- Xu Ding
- Laboratory of Nutrition and Development, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, No. 56 Nan-li-shi Road, Xi-Cheng District, Beijing, 100045, China.
| | - Wei Yang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Qinghua Ren
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Jiajian Hu
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Shen Yang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Wei Han
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Jing Wang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Xu Wang
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Huanmin Wang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
12
|
The Flavone Luteolin Improves Central Nervous System Disorders by Different Mechanisms: A Review. J Mol Neurosci 2018; 65:491-506. [DOI: 10.1007/s12031-018-1094-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/21/2018] [Indexed: 01/17/2023]
|
13
|
Tenner AJ, Stevens B, Woodruff TM. New tricks for an ancient system: Physiological and pathological roles of complement in the CNS. Mol Immunol 2018; 102:3-13. [PMID: 29958698 DOI: 10.1016/j.molimm.2018.06.264] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022]
Abstract
While the mechanisms underlying the functions of the complement system in the central nervous system (CNS) and systemically, namely opsonization, chemotaxis, membrane lysis, and regulation of inflammation are the same, the plethora of functions that complement orchestrates in the central nervous system (CNS) is complex. Strictly controlled expression of complement effector molecules, regulators and receptors across the gamut of life stages (embryogenesis, development and maturation, aging and disease) dictate fascinating contributions for this ancient system. Furthermore, it is becoming apparent that complement functions differ widely across distinct brain regions. This review provides a comprehensive overview of the newly identified roles for complement in the brain, including its roles in CNS development and function, during aging and in the processes of neurodegeneration. The diversity and selectively of beneficial and detrimental activities of complement, while challenging, should lead to precision targeting of specific components to provide disease modifying treatments for devastating psychiatric and neurodegenerative disorders that are still without effective treatment.
Collapse
Affiliation(s)
- Andrea J Tenner
- Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, CA, United States.
| | - Beth Stevens
- F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Department of Neurobiology, Harvard Medical School, Boston, MA, United States; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
14
|
Hussain G, Zhang L, Rasul A, Anwar H, Sohail MU, Razzaq A, Aziz N, Shabbir A, Ali M, Sun T. Role of Plant-Derived Flavonoids and Their Mechanism in Attenuation of Alzheimer's and Parkinson's Diseases: An Update of Recent Data. Molecules 2018; 23:E814. [PMID: 29614843 PMCID: PMC6017497 DOI: 10.3390/molecules23040814] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/25/2018] [Accepted: 03/27/2018] [Indexed: 12/13/2022] Open
Abstract
Neurodegeneration is a progressive loss of neuronal cells in certain regions of the brain. Most of the neurodegenerative disorders (NDDs) share the communal characteristic such as damage or reduction of various cell types typically including astrocytes and microglial activity. Several compounds are being trialed to treat NDDs but they possess solitary symptomatic advantages along with copious side effects. The finding of more enthralling and captivating compounds to suspend and standstill the pathology of NDDs will be considered as a hallmark of present times. Phytochemicals possess the potential to alternate the synthetic line of therapy against NDDs. The present review explores the potential efficacy of plant-derived flavonoids against most common NDDs including Alzheimer's disease (AD) and Parkinson's disease (PD). Flavonoids are biologically active phytochemicals which possess potential pharmacological effects, including antiviral, anti-allergic, antiplatelet, anti-inflammatory, anti-tumor, anti-apoptotic and anti-oxidant effects and are able to attenuate the pathology of various NDDs through down-regulating the nitric oxide (NO) production, by reducing the tumor necrosis factor-α (TNF-α), by reducing the excitotoxicity of superoxide as well as acting as tyrosine kinase (TK) and monoamine oxidase (MAO) inhibiting enzyme.
Collapse
Affiliation(s)
- Ghulam Hussain
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Longbin Zhang
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China.
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Haseeb Anwar
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Muhammad Umar Sohail
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Aroona Razzaq
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Nimra Aziz
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Asghar Shabbir
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad 44000, Pakistan.
| | - Muhammad Ali
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China.
| |
Collapse
|
15
|
Hwang CJ, Lee HP, Choi DY, Jeong HS, Kim TH, Lee TH, Kim YM, Moon DB, Park SS, Kim SY, Oh KW, Hwang DY, Han SB, Lee HJ, Hong JT. Inhibitory effect of thiacremonone on MPTP-induced dopaminergic neurodegeneration through inhibition of p38 activation. Oncotarget 2018; 7:46943-46958. [PMID: 27409674 PMCID: PMC5216915 DOI: 10.18632/oncotarget.10504] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/30/2016] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is implicated for dopaminergic neurodegeneration. Sulfur compounds extracted from garlic have been shown to have anti-inflammatory properties. Previously, we have investigated that thiacremonone, a sulfur compound isolated from garlic has anti-inflammatory effects on several inflammatory disease models. To investigate the protective effect of thiacremonone against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced behavioral impairment and dopaminergic neurodegeneration, 8 week old ICR mice were given thiacremonone (10 mg/kg) in drinking water for 1 month and received intraperitoneal injection of MPTP (15 mg/kg, four times with 2 h interval) during the last 7 days of treatment. Our data showed that thiacremonone decreased MPTP-induced behavioral impairments (Rotarod test, Pole test, and Gait test), dopamine depletion and microglia and astrocytes activations as well as neuroinflammation. Higher activation of p38 was found in the substantia nigra and striatum after MPTP injection, but p38 activation was reduced in thiacremonone treated group. In an in vitro study, thiacremonone (1, 2, and 5 μg/ml) effectively decreased MPP+ (0.5 mM)-induced glial activation, inflammatory mediators generation and dopaminergic neurodegeneration in cultured astrocytes and microglial BV-2 cells. Moreover, treatment of p38 MAPK inhibitor SB203580 (10 μM) further inhibited thiacremonone induced reduction of neurodegeneration and neuroinflammation. These results indicated that the anti-inflammatory compound, thiacremonone, inhibited neuroinflammation and dopaminergic neurodegeneration through inhibition of p38 activation.
Collapse
Affiliation(s)
- Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, Daehak-Ro, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Heon Sang Jeong
- College of Agriculture, Life and Environments Sciences, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Tae Hoon Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Tae Hyung Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Young Min Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Dae Bong Moon
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Sung Sik Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Sun Young Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Ki-Wan Oh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Dae Yeon Hwang
- College of Natural Resources & Life Science, Pusan National University, Pusan, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Hwa-Jeong Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
16
|
Dong L, Hu Y, Zhou L, Cheng X. P2X7 receptor antagonist protects retinal ganglion cells by inhibiting microglial activation in a rat chronic ocular hypertension model. Mol Med Rep 2017; 17:2289-2296. [PMID: 29207073 PMCID: PMC5783460 DOI: 10.3892/mmr.2017.8137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/09/2017] [Indexed: 12/31/2022] Open
Abstract
Microglial activation and the release of pro-inflammatory cytokines occur during early glaucoma. However, the exact mechanism underlying the initiation of the microglial activation process remains unclear. Thus, the present study investigated the potential role of a purine receptor subtype, the P2X purinoceptor 7 (P2X7) receptor, during microglial activation in the retinal tissues of a rat chronic ocular hypertension (COH) model. This was achieved by cauterizing 3 of the 4 episcleral veins. Microglial activation and caspase-1 upregulation were observed in COH rat retinas by immunohistochemical and western blotting techniques. Intravitreal injection of 2′,3′-O-(4-benzoylbenzoyl)-ATP (BzATP), a P2X7 receptor agonist, induced microglial activation in normal rat retinal tissues, which was alleviated by pretreatment with the P2X7 receptor antagonist, Brilliant Blue G (BBG). BBG further attenuated caspase-1 increment in COH rat retinal tissues. The data demonstrated that BBG reduced TUNEL-positive retinal ganglion cells in whole-mount retinal tissues with COH and normal retinal tissues following intravitreal injection with BzATP. One may conclude that the P2X7 receptor may be involved in microglial activation in the COH retina and could be considered a target for neuronal protection in glaucoma.
Collapse
Affiliation(s)
- Lingdan Dong
- Central Laboratory, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Yanhong Hu
- Nursing Department, Medical School of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Long Zhou
- Department of Pathology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Xianglin Cheng
- Department of Neurology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
17
|
Autophagy regulates MAVS signaling activation in a phosphorylation-dependent manner in microglia. Cell Death Differ 2016; 24:276-287. [PMID: 28141795 DOI: 10.1038/cdd.2016.121] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 08/26/2016] [Accepted: 09/19/2016] [Indexed: 11/08/2022] Open
Abstract
Mitochondrial antiviral signaling (MAVS) protein has an important role in antiviral immunity and autoimmunity. However, the pathophysiological role of this signaling pathway, especially in the brain, remains elusive. Here we demonstrated that MAVS signaling existed and mediated poly(I:C)-induced inflammation in the brain. Along with the MAVS signaling activation, there was an induction of autophagic activation. Autophagy negatively regulated the activity of MAVS through direct binding of LC3 to the LIR motif Y(9)xxI(12) of MAVS. We also found that c-Abl kinase phosphorylated MAVS and regulated its interaction with LC3. Interestingly, tyrosine phosphorylation of MAVS was required for downstream signaling activation. Importantly, in vivo data showed that the deficiency of MAVS or c-Abl prevented MPTP-induced microglial activation and dopaminergic neuron loss. Together, our findings reveal the molecular mechanisms underlying the regulation of MAVS-dependent microglial activation in the nervous system, thus providing a potential target for the treatment of microglia-driven inflammatory brain diseases.
Collapse
|
18
|
Anti-Inflammatory Activity of Bee Venom in BV2 Microglial Cells: Mediation of MyD88-Dependent NF-κB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:3704764. [PMID: 27563334 PMCID: PMC4987476 DOI: 10.1155/2016/3704764] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 06/30/2016] [Indexed: 11/18/2022]
Abstract
Bee venom has long been used as a traditional folk medicine in Korea. It has been reportedly used for the treatment of arthritis, cancer, and inflammation. Although its anti-inflammatory activity in lipopolysaccharide- (LPS-) stimulated inflammatory cells has been reported, the exact mechanism of its anti-inflammatory action has not been fully elucidated. Therefore, the aim of this study was to investigate the anti-inflammatory mechanism of bee venom in BV2 microglial cells. We first investigated whether NO production in LPS-activated BV2 cells was inhibited by bee venom, and further iNOS mRNA and protein expressions were determined. The mRNA and protein levels of proinflammatory cytokines were examined using semiquantitative RT-PCR and immunoblotting, respectively. Moreover, modulation of the transcription factor NF-κB by bee venom was also investigated using a luciferase assay. LPS-induced NO production in BV2 microglial cells was significantly inhibited in a concentration-dependent manner upon pretreatment with bee venom. Bee venom markedly reduced the mRNA expression of COX-2, TNF-α, IL-1β, and IL-6 and suppressed LPS-induced activation of MyD88 and IRAK1 and phosphorylation of TAK1. Moreover, NF-κB translocation by IKKα/β phosphorylation and subsequent IκB-α degradation were also attenuated. Thus, collectively, these results indicate that bee venom exerts its anti-inflammatory activity via the IRAK1/TAK1/NF-κB signaling pathway.
Collapse
|
19
|
Aging-related 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurochemial and behavioral deficits and redox dysfunction: improvement by AK-7. Exp Gerontol 2016; 82:19-29. [PMID: 27235848 DOI: 10.1016/j.exger.2016.05.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/09/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023]
Abstract
Aging is a prominent risk factor for the occurrence and progression of Parkinson disease (PD). Aging animals are more significant for PD research than young ones. It is promising to develop effective treatments for PD through modulation of aging-related molecules. Sirtuin 2 (SIRT2), a strong deacetylase highly expressed in the brain, has been implicated in the aging process. In our present study, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP, 12mg/kg once daily) was observed to bring about significant behavioral deficits and striatal dopamine depletion in aging male and female mice, while it did not do so in young animals. MPTP did not cause significant reduction in striatal 5-hydroxytryptamine content in aging male and female mice. Furthermore, we observed that MPTP treatment resulted in significant reduction in GSH content and significant increase in MDA content and SIRT2 expression in the substantia nigra (SN) of aging mice, while it did not do so in young animals. Importantly, we observed that AK-7 (a selective SIRT2 inhibitor) significantly improved behavior abnormality and neurochemical deficits in aging male and female mice treated with MPTP. Significant increase in GSH content and significant decrease in MDA content were also observed in the SN of aging male and female mice co-treated with MPTP and AK-7 compared with the MPTP-treated animals. Our results indicated that MPTP induce aging-related neurochemical and behavioural deficits and dysfunction of redox network in male and female mice and AK-7 may be neuroprotective in PD through modulating redox network.
Collapse
|
20
|
Chen H, Wang X, Wang M, Yang L, Yan Z, Zhang Y, Liu Z. Behavioral and Neurochemical Deficits in Aging Rats with Increased Neonatal Iron Intake: Silibinin's Neuroprotection by Maintaining Redox Balance. Front Aging Neurosci 2015; 7:206. [PMID: 26578951 PMCID: PMC4623400 DOI: 10.3389/fnagi.2015.00206] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 10/12/2015] [Indexed: 02/04/2023] Open
Abstract
Aging is a critical risk factor for Parkinson's disease. Silibinin, a major flavonoid in Silybum marianum, has been suggested to display neuroprotective properties against various neurodegenerative diseases. In the present study, we observed that neonatal iron (120 μg/g body weight) supplementation resulted in significant abnormality of behavior and depletion of striatal dopamine (DA) in the aging male and female rats while it did not do so in the young male and female rats. No significant change in striatal serotonin content was observed in the aging male and female rats with neonatal supplementation of the same dose of iron. Furthermore, we found that the neonatal iron supplementation resulted in significant increase in malondialdehyde (MDA) and decrease in glutathione (GSH) in the substantia nigra (SN) of the aging male and female rats. No significant change in content of MDA and GSH was observed in the cerebellum of the aging male and female rats with the neonatal iron supplementation. Interestingly, silibinin (25 and 50 mg/kg body weight) treatment significantly and dose-dependently attenuated depletion of striatal DA and improved abnormality of behavior in the aging male and female rats with the neonatal iron supplementation. Moreover, silibinin significantly reduced MDA content and increased GSH content in the SN of the aging male and female rats. Taken together, our results indicate that elevated neonatal iron supplementation may result in neurochemical and behavioral deficits in the male and female rats with aging and silibinin may exert dopaminergic neuroprotection by maintaining redox balance.
Collapse
Affiliation(s)
- Hanqing Chen
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai , China ; School of Biotechnology and Food Engineering, Hefei University of Technology , Hefei , China
| | - Xijin Wang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Meihua Wang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Liu Yang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Zhiqiang Yan
- Shanghai Laboratory Animal Center, Chinese Academy of Sciences , Shanghai , China
| | - Yuhong Zhang
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University , Shanghai , China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai , China
| |
Collapse
|
21
|
Weinstein JR, Quan Y, Hanson JF, Colonna L, Iorga M, Honda SI, Shibuya K, Shibuya A, Elkon KB, Möller T. IgM-Dependent Phagocytosis in Microglia Is Mediated by Complement Receptor 3, Not Fcα/μ Receptor. THE JOURNAL OF IMMUNOLOGY 2015; 195:5309-17. [PMID: 26500348 DOI: 10.4049/jimmunol.1401195] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/29/2015] [Indexed: 12/11/2022]
Abstract
Microglia play an important role in receptor-mediated phagocytosis in the CNS. In brain abscess and other CNS infections, invading bacteria undergo opsonization with Igs or complement. Microglia recognize these opsonized pathogens by Fc or complement receptors triggering phagocytosis. In this study, we investigated the role of Fcα/μR, the less-studied receptor for IgM and IgA, in microglial phagocytosis. We showed that primary microglia, as well as N9 microglial cells, express Fcα/μR. We also showed that anti-Staphylococcus aureus IgM markedly increased the rate of microglial S. aureus phagocytosis. To unequivocally test the role of Fcα/μR in IgM-mediated phagocytosis, we performed experiments in microglia from Fcα/μR(-/-) mice. Surprisingly, we found that IgM-dependent phagocytosis of S. aureus was similar in microglia derived from wild-type or Fcα/μR(-/-) mice. We hypothesized that IgM-dependent activation of complement receptors might contribute to the IgM-mediated increase in phagocytosis. To test this, we used immunologic and genetic inactivation of complement receptor 3 components (CD11b and CD18) as well as C3. IgM-, but not IgG-mediated phagocytosis of S. aureus was reduced in wild-type microglia and macrophages following preincubation with an anti-CD11b blocking Ab. IgM-dependent phagocytosis of S. aureus was also reduced in microglia derived from CD18(-/-) and C3(-/-) mice. Taken together, our findings implicate complement receptor 3 and C3, but not Fcα/μR, in IgM-mediated phagocytosis of S. aureus by microglia.
Collapse
Affiliation(s)
- Jonathan R Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195;
| | - Yi Quan
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195
| | - Josiah F Hanson
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195
| | - Lucrezia Colonna
- Division of Rheumatology, Department of Medicine, School of Medicine, University of Washington, Seattle, WA 98195; and
| | - Michael Iorga
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195
| | - Shin-ichiro Honda
- Department of Immunology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Kazuko Shibuya
- Department of Immunology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Akira Shibuya
- Department of Immunology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Keith B Elkon
- Division of Rheumatology, Department of Medicine, School of Medicine, University of Washington, Seattle, WA 98195; and
| | - Thomas Möller
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195
| |
Collapse
|
22
|
Wang X, Guan Q, Wang M, Yang L, Bai J, Yan Z, Zhang Y, Liu Z. Aging-related rotenone-induced neurochemical and behavioral deficits: role of SIRT2 and redox imbalance, and neuroprotection by AK-7. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2553-63. [PMID: 26089639 PMCID: PMC4466888 DOI: 10.2147/dddt.s81539] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aging is one of the strongest risk factors for Parkinson’s disease (PD). SIRT2 has been implicated in the aging process. It is pertinent to investigate the role of SIRT2 in aging-related dopaminergic neurotoxicity and to develop effective therapeutic strategies for PD through the use of aging animals. In this study, we observed that rotenone induced significant behavior abnormality and striatal dopamine depletion in aging rats, while it did not do so in young rats. No significant change in striatal serotonin level was observed in the aging rats after rotenone administration. There was also aging-related rotenone-induced increase in substantia nigra (SN) SIRT2 expression in the rats. In addition, there was aging-related rotenone-induced SN malondialdehyde (MDA) increase and glutathione (GSH) decrease in the rats. No significant changes in cerebellar SIRT2, MDA, or GSH levels were observed in the aging rats after rotenone administration. Striatal dopamine content was significantly inversely correlated with SN SIRT2 expression in the rats. AK-7 significantly diminished striatal dopamine depletion and improved behavior abnormality in the rotenone-treated aging rats. Furthermore, AK-7 significantly decreased MDA content and increased GSH content in the SN of rotenone-treated aging rats. Finally, the effect of AK-7 on dopaminergic neurons and redox imbalance was supported by the results from primary mesencephalic cultures. Our study helps to elucidate the mechanism for the participation of aging in PD and suggests that SN SIRT2 may be involved in PD neurodegeneration, that AK-7 may be neuroprotective in PD, and that maintaining redox balance may be one of the mechanisms underlying neuroprotection by AK-7.
Collapse
Affiliation(s)
- Xijin Wang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Qiang Guan
- Department of Neurology, Tongji Hospital, Tongji University, Shanghai, People's Republic of China
| | - Meihua Wang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Liu Yang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jie Bai
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhiqiang Yan
- Shanghai Laboratory Animal Center, Chinese Academy of Sciences, Tongji University, Shanghai, People's Republic of China
| | - Yuhong Zhang
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
23
|
Wang X, Wang M, Yang L, Bai J, Yan Z, Zhang Y, Liu Z. Inhibition of Sirtuin 2 exerts neuroprotection in aging rats with increased neonatal iron intake. Neural Regen Res 2015; 9:1917-22. [PMID: 25558243 PMCID: PMC4281432 DOI: 10.4103/1673-5374.145361] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2014] [Indexed: 11/21/2022] Open
Abstract
Impaired iron homeostasis may cause damage to dopaminergic neurons and is critically involved in the pathogenesis of Parkinson's disease. At present, very little is understood about the effect of neonatal iron intake on behavior in aging animals. Therefore, we hypothesized that increased neonatal iron intake would result in significant behavior abnormalities and striatal dopamine depletion during aging, and Sirtuin 2 contributes to the age-related neurotoxicity. In the present study, we observed that neonatal iron intake (120 μg/g per day) during postnatal days 10–17 resulted in significant behavior abnormalities and striatal dopamine depletion in aging rats. Furthermore, after AK-7 (a selective Sirtuin 2 inhibitor) was injected into the substantia nigra at postnatal 540 days and 570 days (5 μg/side per day), striatal dopamine depletion was significantly diminished and behavior abnormality was improved in aging rats with neonatal iron intake. Experimental findings suggest that increased neonatal iron intake may result in Parkinson's disease-like neurochemical and behavioral deficits with aging, and inhibition of Sirtuin 2 expression may be a neuroprotective measure in Parkinson's disease.
Collapse
Affiliation(s)
- Xijin Wang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meihua Wang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liu Yang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Bai
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiqiang Yan
- Shanghai Laboratory Animal Center, Chinese Academy of Sciences, Shanghai, China
| | - Yuhong Zhang
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Zhang J, Niu N, Li B, McNutt MA. Neuron-derived IgG protects neurons from complement-dependent cytotoxicity. J Histochem Cytochem 2013; 61:869-79. [PMID: 23979841 DOI: 10.1369/0022155413504196] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Passive immunity of the nervous system has traditionally been thought to be predominantly due to the blood-brain barrier. This concept must now be revisited based on the existence of neuron-derived IgG. The conventional concept is that IgG is produced solely by mature B lymphocytes, but it has now been found to be synthesized by murine and human neurons. However, the function of this endogenous IgG is poorly understood. In this study, we confirm IgG production by rat cortical neurons at the protein and mRNA levels, with 69.0 ± 5.8% of cortical neurons IgG-positive. Injury to primary-culture neurons was induced by complement leading to increases in IgG production. Blockage of neuron-derived IgG resulted in more neuronal death and early apoptosis in the presence of complement. In addition, FcγRI was found in microglia and astrocytes. Expression of FcγR I in microglia was increased by exposure to neuron-derived IgG. Release of NO from microglia triggered by complement was attenuated by neuron-derived IgG, and this attenuation could be reversed by IgG neutralization. These data demonstrate that neuron-derived IgG is protective of neurons against injury induced by complement and microglial activation. IgG appears to play an important role in maintaining the stability of the nervous system.
Collapse
|
25
|
Yang F, Wu L, Guo X, Wang D, Li Y. Improved retinal ganglion cell survival through retinal microglia suppression by a chinese herb extract, triptolide, in the DBA/2J mouse model of glaucoma. Ocul Immunol Inflamm 2013; 21:378-89. [PMID: 23876132 DOI: 10.3109/09273948.2013.806989] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
To investigate the changes in retinal microglia and retinal ganglion cell (RGC) survival after long-term administration of a Chinese herb extract, triptolide, in a DBA/2J mice. DBA/2J mice (n = 96) were administered triptolide (n = 48) 25 µg/kg or vehicle (n = 48) and were judged at 7, 9, 11 months of age. Long-term triptolide treatment tended to attenuate the anterior segment pathology in experimental group, though intraocular pressure was not significantly different between the two groups. In the experimental group, RGC survival was improved (7, 9, 11 months: p = 0.035, 0.004, 0.014), and microglia activation was suppressed based on a more ramified appearance (9, 11 months: p = 0.024, 0.013) and a lower total microglial cell count (7, 9, 11 months: p = 0.028, 0.025, 0.014). Double-immunofluorescence staining revealed TNF? localized to microglia, TNFR1 localized to the RGCs and nerve fiber layer. These findings indicate that long-term triptolide administration suppressed microglia activation and improved RGC survival in DBA/2J mice.
Collapse
Affiliation(s)
- Fan Yang
- Peking university third hospital , Beijing , China
| | | | | | | | | |
Collapse
|
26
|
Zhang J, Niu N, Wang M, McNutt MA, Zhang D, Zhang B, Lu S, Liu Y, Liu Z. Neuron-derived IgG protects dopaminergic neurons from insult by 6-OHDA and activates microglia through the FcγR I and TLR4 pathways. Int J Biochem Cell Biol 2013; 45:1911-20. [PMID: 23791745 DOI: 10.1016/j.biocel.2013.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/24/2013] [Accepted: 06/03/2013] [Indexed: 12/09/2022]
Abstract
Oxidative and immune attacks from the environment or microglia have been implicated in the loss of dopaminergic neurons of Parkinson's disease. The role of IgG which is an important immunologic molecule in the process of Parkinson's disease has been unclear. Evidence suggests that IgG can be produced by neurons in addition to its traditionally recognized source B lymphocytes, but its function in neurons is poorly understood. In this study, extensive expression of neuron-derived IgG was demonstrated in dopaminergic neurons of human and rat mesencephalon. With an in vitro Parkinson's disease model, we found that neuron-derived IgG can improve the survival and reduce apoptosis of dopaminergic neurons induced by 6-hydroxydopamine toxicity, and also depress the release of NO from microglia triggered by 6-hydroxydopamine. Expression of TNF-α and IL-10 in microglia was elevated to protective levels by neuron-derived IgG at a physiologic level via the FcγR I and TLR4 pathways and microglial activation could be attenuated by IgG blocking. All these data suggested that neuron-derived IgG may exert a self-protective function by activating microglia properly, and IgG may be involved in maintaining immunity homeostasis in the central nervous system and serve as an active factor under pathological conditions such as Parkinson's disease.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Human Anatomy, Weifang Medical University, Weifang 261053, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sanchez-Guajardo V, Barnum C, Tansey M, Romero-Ramos M. Neuroimmunological processes in Parkinson's disease and their relation to α-synuclein: microglia as the referee between neuronal processes and peripheral immunity. ASN Neuro 2013; 5:113-39. [PMID: 23506036 PMCID: PMC3639751 DOI: 10.1042/an20120066] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 03/15/2013] [Accepted: 03/19/2013] [Indexed: 12/15/2022] Open
Abstract
The role of neuroinflammation and the adaptive immune system in PD (Parkinson's disease) has been the subject of intense investigation in recent years, both in animal models of parkinsonism and in post-mortem PD brains. However, how these processes relate to and modulate α-syn (α-synuclein) pathology and microglia activation is still poorly understood. Specifically, how the peripheral immune system interacts, regulates and/or is induced by neuroinflammatory processes taking place during PD is still undetermined. We present herein a comprehensive review of the features and impact that neuroinflamation has on neurodegeneration in different animal models of nigral cell death, how this neuroinflammation relates to microglia activation and the way microglia respond to α-syn in vivo. We also discuss a possible role for the peripheral immune system in animal models of parkinsonism, how these findings relate to the state of microglia activation observed in these animal models and how these findings compare with what has been observed in humans with PD. Together, the available data points to the need for development of dual therapeutic strategies that modulate microglia activation to change not only the way microglia interact with the peripheral immune system, but also to modulate the manner in which microglia respond to encounters with α-syn. Lastly, we discuss the immune-modulatory strategies currently under investigation in animal models of parkinsonism and the degree to which one might expect their outcomes to translate faithfully to a clinical setting.
Collapse
Key Words
- lymphocytes
- m1/m2 phenotype
- microglia
- neuroinflammation
- parkinson’s disease
- α-synuclein
- 6-ohda, 6-hydroxydopamine
- ad, alzheimer’s disease
- apc, antigen-presenting cell
- α-syn, α-synuclein
- bbb, brain–blood barrier
- bcg, bacille calmette–guérin
- bm, bone marrow
- cfa, complete freund’s adjuvant
- cm, conditioned media
- cns, central nervous system
- cox, cyclooxygenase
- cr, complement receptor
- csf, cerebrospinal fluid
- da, dopamine
- eae, experimental autoimmune encephalomyelitis
- ga, galatiramer acetate
- gdnf, glial-derived neurotrophic factor
- gfp, green fluorescent protein
- hla-dr, human leucocyte antigen type dr
- ifnγ, interferon γ
- igg, immunoglobulin g
- il, interleukin
- inos, inducible nitric oxide synthase
- lamp, lysosome-associated membrane protein
- lb, lewy body
- lps, lipopolysaccharide
- mhc, major histocompatibility complex
- mptp, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- nfκb, nuclear factor κb
- nk, natural killer
- no, nitric oxide
- pd, parkinson’s disease
- pet, positron-emission tomography
- prp, prion protein
- raav, recombinant adeno-associated virus
- rns, reactive nitrogen species
- ros, reactive oxygen species
- sn, substantia nigra
- snp, single nucleotide polymorphism
- tcr, t-cell receptor
- tgfβ, tumour growth factor β
- th, tyrosine hydroxylase
- th1, t helper 1
- tlr, toll-like receptor
- tnf, tumour necrosis factor
- treg, regulatory t-cell
- vip, vasoactive intestinal peptide
- wt, wild-type
Collapse
Affiliation(s)
- Vanesa Sanchez-Guajardo
- *CNS Disease Modeling Group, Department of Biomedicine, Ole Worms Allé 3,
Aarhus University, DK-8000 Aarhus C, Denmark
| | - Christopher J. Barnum
- †Department of Physiology, Emory University, School of Medicine, Atlanta, GA
30233, U.S.A
| | - Malú G. Tansey
- †Department of Physiology, Emory University, School of Medicine, Atlanta, GA
30233, U.S.A
| | - Marina Romero-Ramos
- *CNS Disease Modeling Group, Department of Biomedicine, Ole Worms Allé 3,
Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
28
|
Lin YP, Chen TY, Tseng HW, Lee MH, Chen ST. Chemical and biological evaluation of nephrocizin in protecting nerve growth factor-differentiated PC12 cells by 6-hydroxydopamine-induced neurotoxicity. PHYTOCHEMISTRY 2012; 84:102-115. [PMID: 22954731 DOI: 10.1016/j.phytochem.2012.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 07/12/2012] [Accepted: 07/25/2012] [Indexed: 06/01/2023]
Abstract
The neurotoxin 6-hydroxydopamine (6-OHDA) has been widely used to generate an experimental model of Parkinson's disease. This model is crucial in the search for compounds that diminish 6-OHDA-induced nerve growth factor (NGF)-differentiated PC12 cell death. Nephrocizin (luteolin-7-O-β-D-glucopyranoside), a flavone glycoside, was isolated from widely distributed plants. The protective effects of pre-treatment with nephrocizin on the induced neurotoxicity in PC12 cells by 6-OHDA and its oxidative products, H₂O₂-, and p-quinone, were evaluated herein. Nephrocizin promoted cell viability, scavenged ROS-related products, increased cellular glutathione (GSH) levels, and reduced caspase-3 and -8 activities in 6-OHDA-, H₂O₂-, or p-quinone-treated PC12 cells. Furthermore, nephrocizin-conjugated metabolites in PC12 cells were identified with the boronate-affinity method and LC-MS technology, and preferential regioselectivity at the C2' and C5' positions by the nephrocizin-GSH (or NAC) adduct method was observed. These lines of evidence established that nephrocizin could form a dimer to diminish the intracellular ROS. These results demonstrate the first neuroprotective mechanism of nephrocizin against 6-OHDA-, H₂O₂- or p-quinone-induced cytotoxicity in PC12 cells via chemical and biological studies. These dietary antioxidants are potential candidates for use in intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Pei Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan, ROC
| | | | | | | | | |
Collapse
|
29
|
Brennan FH, Anderson AJ, Taylor SM, Woodruff TM, Ruitenberg MJ. Complement activation in the injured central nervous system: another dual-edged sword? J Neuroinflammation 2012; 9:137. [PMID: 22721265 PMCID: PMC3464784 DOI: 10.1186/1742-2094-9-137] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 06/21/2012] [Indexed: 11/28/2022] Open
Abstract
The complement system, a major component of the innate immune system, is becoming increasingly recognised as a key participant in physiology and disease. The awareness that immunological mediators support various aspects of both normal central nervous system (CNS) function and pathology has led to a renaissance of complement research in neuroscience. Various studies have revealed particularly novel findings on the wide-ranging involvement of complement in neural development, synapse elimination and maturation of neural networks, as well as the progression of pathology in a range of chronic neurodegenerative disorders, and more recently, neurotraumatic events, where rapid disruption of neuronal homeostasis potently triggers complement activation. The purpose of this review is to summarise recent findings on complement activation and acquired brain or spinal cord injury, i.e. ischaemic-reperfusion injury or stroke, traumatic brain injury (TBI) and spinal cord injury (SCI), highlighting the potential for complement-targeted therapeutics to alleviate the devastating consequences of these neurological conditions.
Collapse
Affiliation(s)
- Faith H Brennan
- The University of Queensland, School of Biomedical Sciences, St Lucia, Brisbane, QLD 4072, Australia
| | | | | | | | | |
Collapse
|
30
|
Wang XJ, Zhang S, Yan ZQ, Zhao YX, Zhou HY, Wang Y, Lu GQ, Zhang JD. Impaired CD200-CD200R-mediated microglia silencing enhances midbrain dopaminergic neurodegeneration: roles of aging, superoxide, NADPH oxidase, and p38 MAPK. Free Radic Biol Med 2011; 50:1094-106. [PMID: 21295135 DOI: 10.1016/j.freeradbiomed.2011.01.032] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 01/24/2011] [Accepted: 01/25/2011] [Indexed: 11/24/2022]
Abstract
CD200-CD200R signaling holds microglia in a quiescent state. Parkinson disease (PD) neurodegeneration may be associated with impairment of CD200-CD200R-mediated microglia silencing in the substantia nigra (SN). In this study, an anti-CD200R blocking antibody (ACDR) selectively and significantly enhanced the susceptibility of dopaminergic neurons to neurotoxicity induced by rotenone (Rot) and iron (Ir) in mesencephalic neuron/glia cultures. Microglia were shown to mediate dopaminergic neurotoxicity induced by ACDR/Rot (combination of ACDR and Rot) and ACDR/Ir (combination of ACDR and Ir). ACDR significantly enhanced the microglial activation induced by Rot and Ir in neuron/glia cultures. NADPH oxidase-mediated superoxide generation was a key contributor to dopaminergic neurotoxicity induced by ACDR/Rot and ACDR/Ir. p38 MAPK contributed to NADPH oxidase activation induced by ACDR/Rot and ACDR/Ir. Interestingly, there were a decrease in CD200 expression (mRNA and protein) and an enhancement of microglial response (MHCII mRNA and ICAM-1 protein) in the rat SN with aging. ICAM-1 expression was significantly inversely correlated with CD200 expression. These results strongly indicate the participation of SN CD200-CD200R dysfunction in the etiopathogenesis of PD and provide a new insight into the molecular mechanisms underlying the involvement of aging in PD and help to elucidate the mechanisms of the combined involvement of immune/inflammatory factors, environmental substances, and aging in PD.
Collapse
Affiliation(s)
- Xi-Jin Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
In view of the population-specific heterogeneity in reported genetic risk factors for Parkinson's disease (PD), we conducted a genome-wide association study (GWAS) in a large sample of PD cases and controls from the Netherlands. After quality control (QC), a total of 514,799 SNPs genotyped in 772 PD cases and 2024 controls were included in our analyses. Direct replication of SNPs within SNCA and BST1 confirmed these two genes to be associated with PD in the Netherlands (SNCA, rs2736990: P = 1.63 × 10(-5), OR = 1.325 and BST1, rs12502586: P = 1.63 × 10(-3), OR = 1.337). Within SNCA, two independent signals in two different linkage disequilibrium (LD) blocks in the 3' and 5' ends of the gene were detected. Besides, post-hoc analysis confirmed GAK/DGKQ, HLA and MAPT as PD risk loci among the Dutch (GAK/DGKQ, rs2242235: P = 1.22 × 10(-4), OR = 1.51; HLA, rs4248166: P = 4.39 × 10(-5), OR = 1.36; and MAPT, rs3785880: P = 1.9 × 10(-3), OR = 1.19).
Collapse
|
32
|
Członkowska A, Kurkowska-Jastrzębska I. Inflammation and gliosis in neurological diseases--clinical implications. J Neuroimmunol 2010; 231:78-85. [PMID: 20943275 DOI: 10.1016/j.jneuroim.2010.09.020] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The inflammatory reaction accompany all acute processes in the central nervous system (CNS), (as stroke or traumatic brain injury) and chronic neurodegenerative processes (as Parkinson's or Alzheimer's disease), and through the stage of cleaning of damage tissue, contribute to recovery and regeneration and eventually to restoration of the function. However many studies showed that inflammation in the CNS may be harmful because of an excessive vulnerability of the nervous tissue or impaired regulation. Manipulation of the inflammation is now one of the approaches in the treatment of the various diseases of the CNS.
Collapse
Affiliation(s)
- Anna Członkowska
- 2nd Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland.
| | | |
Collapse
|
33
|
Altered regulation of CD200 receptor in monocyte-derived macrophages from individuals with Parkinson's disease. Neurochem Res 2010; 35:540-7. [PMID: 19924532 DOI: 10.1007/s11064-009-0094-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2009] [Indexed: 10/20/2022]
Abstract
Microglia are the representative myeloid cells in the brain, and their over-activation plays an important role in the pathogenesis of Parkinson's disease (PD). Microglia activation is believed to be regulated by the CD200-CD200R signaling. As the peripheral counterpart of microglia, monocyte-derived macrophages (MDMs) share the same progenitor and antigen markers, and they have similar biological behaviors and mirror microglial function in the brain. Here, we studied CD200R expression and its regulation in MDMs from 32 PD cases, 27 age-matched old controls, and 28 young controls. We found that the basal CD200R expression is similar in MDMs from young control, old control and PD patients. However, the induction of CD200R expression in MDMs under various conditions is impaired in the old groups, especially in PD patients. There was a selective decrease in CD200R expression induced by co-culture with dying PC12 cells in MDMs from PD cases, as compared with MDMs from the age-matched controls. We also found that the inducible CD200R expression correlated inversely with the onset age of PD and to tumor necrosis factor-alpha (TNF-alpha) released from MDMs. These results suggest an intrinsic abnormality in the CD200-CD200R signaling in MDMs during aging and, especially, in PD. We speculate that in the PD brain,microglia might undergo abnormalities similar to MDMs.
Collapse
|
34
|
Cristóvão AC, Saavedra A, Fonseca CP, Campos F, Duarte EP, Baltazar G. Microglia of rat ventral midbrain recovers its resting state over time in vitro: let microglia rest before work. J Neurosci Res 2010; 88:552-62. [PMID: 19739250 DOI: 10.1002/jnr.22219] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cortical or total brain cultures of microglia are commonly used as a model to study the inflammatory processes in Parkinson's disease. Here we characterize microglia cultures from rat ventral midbrain and evaluate their response to zymosan A. We used specific markers of microglia and evaluated the morphology, the phagocytic activity and reactive oxygen species (ROS) levels of the cells. During the first 10 days in vitro (DIV), cultures presented predominantly cells with a round morphology, expressing CD68 and with high phagocytic activity and ROS production. After 13 DIV, this tendency was reversed, with cultures showing higher number of ramified cells and fewer CD68(+) cells along with lower phagocytic and ROS production capability, suggesting that microglia must be kept in vitro for at least 13 days to recover its resting state. The exposure of cultures with less than 10 DIV to zymosan A significantly decreased cell viability. Exposure of cultures with 13 DIV to zymosan A (0.05, 0.5, or 5 microg/ml) increased the total cell number, the percentage of CD68(+) cells, and the phagocytic activity. Concentrations of zymosan A higher than 5 microg/ml were also effective in activating microglia but significantly decreased the number of viable cells. In summary, microglial cells remain in the activated state for several days after the isolation process and, thus, stimulation of microglia recently isolated can compromise interpretation of the results. However, upon 13 DIV, cells achieve properties of nonactivated microglia and present a characteristic response to a proinflammatory agent.
Collapse
Affiliation(s)
- Ana Clara Cristóvão
- Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | | | | | | | | | | |
Collapse
|
35
|
Luo XG, Ding JQ, Chen SD. Microglia in the aging brain: relevance to neurodegeneration. Mol Neurodegener 2010; 5:12. [PMID: 20334662 PMCID: PMC2852379 DOI: 10.1186/1750-1326-5-12] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Accepted: 03/24/2010] [Indexed: 12/27/2022] Open
Abstract
Microglia cells are the brain counterpart of macrophages and function as the first defense in the brain. Although they are neuroprotective in the young brain, microglia cells may be primed to react abnormally to stimuli in the aged brain and to become neurotoxic and destructive during neurodegeneration. Aging-induced immune senescence occurs in the brain as age-associated microglia senescence, which renders microglia to function abnormally and may eventually promote neurodegeneration. Microglia senescence is manifested by both morphological changes and alterations in immunophenotypic expression and inflammatory profile. These changes are likely caused by microinvironmental factors, but intrinsic factors cannot yet be completely excluded. Microglia senescence appears to underlie the switching of microglia from neuroprotective in the young brain to neurotoxic in the aged brain. The hypothesis of microglia senescence during aging offers a novel perspective on their roles in aging-related neurodegeneration. In Parkinson's disease and Alzheimer's disease, over-activation of microglia may play an active role in the pathogenesis because microglia senescence primes them to be neurotoxic during the development of the diseases.
Collapse
Affiliation(s)
- Xiao-Guang Luo
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | | | | |
Collapse
|
36
|
Li W, Gao G, Guo Q, Jia D, Wang J, Wang X, He S, Liang Q. Function and phenotype of microglia are determined by toll-like receptor 2/toll-like receptor 4 activation sequence. DNA Cell Biol 2009; 28:493-9. [PMID: 19622038 DOI: 10.1089/dna.2009.0856] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Toll-like receptors (TLRs) initiate and maintain host defenses. These receptors play important roles in innate immunity and in various diseases. Different TLRs bind to diverse ligands that trigger distinct protein expression patterns. Few studies have focused on the interaction between different TLRs. We found that TLR2 priming downregulates TLR4 transcription, and expression of TLR4 activation induced major histocompatibility complex II (MHC II), adhesion molecule intercellular adhesion molecule-1 (ICAM-1), phagocytosis marker CD11b/CD18, and Fcgamma receptor (FcgammaR) expression. In contrast, TLR4 priming increases TLR2 transcription and expression. In addition, TLR4 priming increases secretion of certain proinflammatory mediators. Expression of costimulatory molecules CD80/CD86 increases with TLR2 or TLR4 activation sequences. Our results reveal that TLR2/TLR4 activation may determine disease pathogenesis and prognosis.
Collapse
Affiliation(s)
- Weixin Li
- Neurosurgical Department,Tangdu Hospital, Fourth Military Medical University , Xi'an, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Stone DK, Reynolds AD, Mosley RL, Gendelman HE. Innate and adaptive immunity for the pathobiology of Parkinson's disease. Antioxid Redox Signal 2009; 11:2151-66. [PMID: 19243239 PMCID: PMC2788126 DOI: 10.1089/ars.2009.2460] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Innate and adaptive immunity affect the pathogenesis of Parkinson's disease (PD). In particular, activation of microglia influences degeneration of dopaminergic neurons. Cell-to-cell interactions and immune regulation critical for neuronal homeostasis also influence immune responses. The links between T cell immunity and nigrostriatal degeneration are supported by laboratory, animal model, and human pathologic investigations. Immune-associated biomarkers in spinal fluids and brain tissue of patients with idiopathic or familial forms of PD provide means to improve diagnosis and therapeutic monitoring. Relationships between oxidative stress, inflammation, and immune-mediated cell death pathways are examined in this review as they are linked to PD pathogenesis. Harnessing the immune system by drugs or by vaccination remain promising future therapeutic options.
Collapse
Affiliation(s)
- David K Stone
- Center for Neurovirology and Neurodegenerative Disorders, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198-5880, USA
| | | | | | | |
Collapse
|
38
|
Dietert RR. Developmental immunotoxicology: focus on health risks. Chem Res Toxicol 2009; 22:17-23. [PMID: 18783253 DOI: 10.1021/tx800198m] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Developmental immunotoxicity (DIT) has gained attention with the recognition that many chronic diseases of increasing incidence feature immune dysfunction as a component of the disease. The maturing immune system represents a vulnerable target for toxicants as it progresses through a series of novel prenatal and perinatal events that are critical for later-life host defense against a wide array of diseases. These critical maturational windows display a particular sensitivity to chemical disruption with the outcome usually taking the form of persistent immune dysfunction and/or misregulation. For this reason, health risks are significantly increased following early life vs adult immunotoxic exposure. Additionally, DIT-associated health risks are not readily predicted when based on adult-exposure safety data or via the evaluation of an unchallenged immune system in developmental toxicity testing. The same toxicant [e.g., heavy metals, 2,3,7,8-tetraclorodibenzo-p-dioxin (TCDD)] may disrupt different immune maturational processes depending upon the specific developmental timing of exposure and the target organ dose at a given stage of development. Therefore, a single toxicant may promote different immune-associated diseases that are dependent upon the specific window of early life exposure, the gender of the exposed offspring, and the genetic background of the offspring. This perspective considers the linkage between early life chemical exposure, DIT, and the postnatal immune dysfunctions associated with a variety of childhood and adult diseases. Because DIT is linked to a majority of the most significant childhood chronic diseases, safety testing for DIT is a pivotal issue in the protection of children's health.
Collapse
Affiliation(s)
- Rodney R Dietert
- Department of Microbiology and Immunology, C5-135 VMC, College of Veterinary Medicine, Cornell UniVersity, North Tower Road, Ithaca, New York 14853, USA.
| |
Collapse
|
39
|
Luteolin protects dopaminergic neurons from inflammation-induced injury through inhibition of microglial activation. Neurosci Lett 2008; 448:175-9. [DOI: 10.1016/j.neulet.2008.10.046] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Accepted: 10/15/2008] [Indexed: 11/18/2022]
|
40
|
Chen HQ, Wang XJ, Jin ZY, Xu XM, Zhao JW, Xie ZJ. Protective effect of isoflavones from Trifolium pratense on dopaminergic neurons. Neurosci Res 2008; 62:123-30. [DOI: 10.1016/j.neures.2008.07.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2008] [Revised: 06/23/2008] [Accepted: 07/07/2008] [Indexed: 11/27/2022]
|
41
|
Kraneveld AD, Rijnierse A, Nijkamp FP, Garssen J. Neuro-immune interactions in inflammatory bowel disease and irritable bowel syndrome: future therapeutic targets. Eur J Pharmacol 2008; 585:361-74. [PMID: 18417115 DOI: 10.1016/j.ejphar.2008.02.095] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 02/11/2008] [Accepted: 02/20/2008] [Indexed: 12/22/2022]
Abstract
The gastro-intestinal tract is well known for its largest neural network outside the central nervous system and for the most extensive immune system in the body. Research in neurogastroenterology implicates the involvement of both enteric nervous system and immune system in symptoms of inflammatory bowel disease and irritable bowel syndrome. Since both disorders are associated with increased immune cell numbers, nerve growth and activation of both immune cells and nerves, we focus in this review on the involvement of immune cell-nerve interactions in inflammatory bowel disease and irritable bowel syndrome. Firstly, the possible effects of enteric nerves, especially of the nonadrenergic and noncholinergic nerves, on the intestinal immune system and their possible role in the pathogenesis of chronic intestinal inflammatory diseases are described. Secondly, the possible effects of immunological factors, from the innate (chemokines and Toll-like receptors) as well as the adaptive (cytokines and immunoglobulins) immune system, on gastro-intestinal nerves and its potential role in the development of inflammatory bowel disease and irritable bowel syndrome are reviewed. Investigations of receptor-mediated and intracellular signal pathways in neuro-immune interactions might help to develop more effective therapeutic approaches for chronic inflammatory intestinal diseases.
Collapse
Affiliation(s)
- Aletta D Kraneveld
- Division Pharmacology and Pathophysiology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, PO box 80082, 3508 TB Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
42
|
Bouchez G, Sensebé L, Vourc'h P, Garreau L, Bodard S, Rico A, Guilloteau D, Charbord P, Besnard JC, Chalon S. Partial recovery of dopaminergic pathway after graft of adult mesenchymal stem cells in a rat model of Parkinson's disease. Neurochem Int 2008; 52:1332-42. [PMID: 18372079 DOI: 10.1016/j.neuint.2008.02.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 02/06/2008] [Indexed: 11/29/2022]
Abstract
Cellular therapy with adult stem cells appears as an opportunity for treatment of Parkinson's disease. To validate this approach, we studied the effects of transplantation of rat adult bone-marrow mesenchymal stem cells in a rat model of Parkinson's disease. Animals were unilaterally lesioned in the striatum with 6-hydroxydopamine. Two weeks later, group I did not undergo grafting, group II underwent sham grafting, group III was intra-striatal grafted with cells cultured in an enriched medium and group IV was intra-striatal grafted with cells cultured in a standard medium. Rotational amphetamine-induced behavior was measured weekly until animals were killed 6 weeks later. One week after graft, the number of rotations/min was stably decreased by 50% in groups III and IV as compared with groups I and II. At 8 weeks post-lesion, the density of dopaminergic markers in the nerve terminals and cell bodies, i.e. immunoreactive tyrosine hydroxylase, membrane dopamine transporter and vesicular monoamine transporter-2 was significantly higher in group III as compared with group I. Moreover, using microdialysis studies, we observed that while the rate of pharmacologically induced release of dopamine was significantly reduced in lesioned versus intact striatum in no grafted rats, it was similar in both sides in animals transplanted with mesemchymal stem cells. These data demonstrate that graft of adult mesemchymal stem cells reduces behavioral effects induced by 6-hydroxydopamine lesion and partially restores the dopaminergic markers and vesicular striatal pool of dopamine. This cellular approach might be a restorative therapy in Parkinson's disease.
Collapse
Affiliation(s)
- Gaëlle Bouchez
- INSERM U930, Laboratory Biophysique médicale & pharmaceutique, UFR Pharmacie, 31 avenue Monge, Tours, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Beck A, Penner R, Fleig A. Lipopolysaccharide-induced down-regulation of Ca2+ release-activated Ca2+ currents (I CRAC) but not Ca2+-activated TRPM4-like currents (I CAN) in cultured mouse microglial cells. J Physiol 2007; 586:427-39. [PMID: 17991695 DOI: 10.1113/jphysiol.2007.145151] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Microglia are the main immunocompetent cells of the mammalian central nervous system (CNS). Activation of cultured microglial cells and subsequent release of nitric oxide and cytokines critically depends on intracellular calcium levels. Since microglia undergo dramatic morphological, biochemical and electrophysiological changes in response to pathological events in the CNS, we investigated temporal changes in expression levels of ion channels involved in cellular calcium homeostasis in mouse cortical microglial cells in culture. Specifically, we assessed the inward and delayed outward rectifier potassium currents (I IRK and I DRK), calcium (Ca2+) release-activated Ca2+ currents (I CRAC) and Ca2+-activated TRPM4-like currents (I CAN) in non-activated microglia and cells that were activated by exposure to lipopolysaccharide (LPS) between 3 and 48 h. Unstimulated microglial cells, subcultured from an astrocyte coculture, typically exhibited a ramified, rod-shaped morphology. During the first 3 days of culture cell size and shape were maintained, but the percentage of cells showing prominent I IRK went up and those expressing I DRK went down. Cells retaining I DRK exhibited smaller amplitudes, whereas those of I IRK and I CRAC were not affected. However, after 24 h of exposure to 1 microg ml(-1) LPS, most cells showed an amoeboid ('fried egg'-shaped) morphology with a 62% increase in cell capacitance. At that point in time, only 14% of the cells revealed I IRK and 3% had I DRK exclusively, whereas the majority of cells expressed both currents. The amplitudes of I CRAC and I IRK progressively decreased after stimulation, whereas I DRK transiently reached a maximum after 6 h of LPS exposure and then returned to pre-stimulation expression levels. Cultured microglia also revealed TRPM4-like, Ca2+-activated non-selective currents (I CAN) with an EC50 of 1.2 microm [Ca2+]i. The expression levels of this current did not change significantly during and after 24 h of LPS exposure. We propose that LPS-induced down-regulation of I IRK and I CRAC will reduce the cell's capacity to produce significant calcium influx upon receptor activation and result in decreased sensitivity to exogenous stimulation. In this scenario, I CAN expression would remain constant, although its activity would automatically be reduced due to the diminished calcium influx capacity of the cell.
Collapse
Affiliation(s)
- Andreas Beck
- Queen's Center for Biomedical Research, Laboratory of Cell and Molecular Signalling, The Queen's Medical Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| | | | | |
Collapse
|
45
|
Wang XJ, Liu WG, Zhang YH, Lu GQ, Chen SD. Effect of transplantation of c17.2 cells transfected with interleukin-10 gene on intracerebral immune response in rat model of Parkinson's disease. Neurosci Lett 2007; 423:95-9. [PMID: 17673368 DOI: 10.1016/j.neulet.2007.06.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Revised: 06/15/2007] [Accepted: 06/18/2007] [Indexed: 01/18/2023]
Abstract
Currently, regulation of immune response after grafting has become a hot topic in Parkinson's disease (PD) transplantation research. Interleukin-10 (IL-10) is an important regulator of immune system. Presently, we transplanted c17.2 neural stem cells transfected with pcDNA3.1-Hygro-IL-10 vector (IL-10-c17.2 cells) or Mock-c17.2 cells (c17.2 cells transfected with pcDNA3.1-Hygro vector) into the brains of 6-hydroxydopamine-lesioned PD model rats. From days 10 to 60 after grafting, double immunohistochemistry showed that IL-10 expression was detected in IL-10-c17.2 cells in vivo. Further immunohistochemistry analyses revealed that intracerebral cellular (ED1 and CD8) and humoral (C3 and IgM) immune responses were down-regulated in the rats treated with IL-10-c17.2 cells compared with controls treated with Mock-c17.2 cells. The reduction in ED1 immunostaining in the rats treated with IL-10-c17.2 cells remained significant until day 60 after transplantation. Our results suggest the potential application value of IL-10 in the transplantation treatment of PD.
Collapse
Affiliation(s)
- Xi-Jin Wang
- Department of Neurology & Neuroscience Institute, Rui-Jin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | | | | | | | | |
Collapse
|
46
|
Wang XJ, Ye M, Zhang YH, Chen SD. CD200–CD200R Regulation of Microglia Activation in the Pathogenesis of Parkinson’s Disease. J Neuroimmune Pharmacol 2007; 2:259-64. [DOI: 10.1007/s11481-007-9075-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2007] [Accepted: 04/20/2007] [Indexed: 01/11/2023]
|
47
|
Chen HQ, Jin ZY, Li GH. Biochanin A protects dopaminergic neurons against lipopolysaccharide-induced damage through inhibition of microglia activation and proinflammatory factors generation. Neurosci Lett 2007; 417:112-7. [PMID: 17399896 DOI: 10.1016/j.neulet.2006.11.045] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Accepted: 11/27/2006] [Indexed: 10/23/2022]
Abstract
Activation of microglia and consequent release of proinflammatory factors, are believed to contribute to neurodegeneration in Parkinson's disease (PD). Hence, identification of compounds that prevent microglial activation is highly desirable in the search for therapeutic agents for inflammation-mediated neurodegenerative diseases. In this study, we reported that biochanin A, one of the predominant isoflavones in Trifolium pratense, attenuated lipopolysaccharide (LPS)-induced decrease in dopamine uptake and the number of dopaminergic neurons in a dose-dependent manner in rat mesencephalic neuron-glia cultures. Moreover, biochanin A also significantly inhibited LPS-induced activation of microglia and production of tumor necrosis factor-alpha, nitric oxide and superoxide in mesencephalic neuron-glia cultures and microglia-enriched cultures. This study suggested for the first time that biochanin A protected dopaminergic neurons against LPS-induced damage through inhibition of microglia activation and proinflammatory factors generation.
Collapse
Affiliation(s)
- Han-Qing Chen
- School of Food Science and Technology, Southern Yangtze University, 1800 Lihu Road, Wuxi, Jiangsu 214122, P.R. China
| | | | | |
Collapse
|