1
|
Luo W, Xu C, Li L, Ji Y, Wang Y, Li Y, Ye Y. Perfluoropentane-based oxygen-loaded nanodroplets reduce microglial activation through metabolic reprogramming. Neural Regen Res 2025; 20:1178-1191. [PMID: 38989955 PMCID: PMC11438333 DOI: 10.4103/nrr.nrr-d-23-01299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/05/2024] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00032/figure1/v/2024-07-06T104127Z/r/image-tiff Microglia, the primary immune cells within the brain, have gained recognition as a promising therapeutic target for managing neurodegenerative diseases within the central nervous system, including Parkinson's disease. Nanoscale perfluorocarbon droplets have been reported to not only possess a high oxygen-carrying capacity, but also exhibit remarkable anti-inflammatory properties. However, the role of perfluoropentane in microglia-mediated central inflammatory reactions remains poorly understood. In this study, we developed perfluoropentane-based oxygen-loaded nanodroplets (PFP-OLNDs) and found that pretreatment with these droplets suppressed the lipopolysaccharide-induced activation of M1-type microglia in vitro and in vivo, and suppressed microglial activation in a mouse model of Parkinson's disease. Microglial suppression led to a reduction in the inflammatory response, oxidative stress, and cell migration capacity in vitro. Consequently, the neurotoxic effects were mitigated, which alleviated neuronal degeneration. Additionally, ultrahigh-performance liquid chromatography-tandem mass spectrometry showed that the anti-inflammatory effects of PFP-OLNDs mainly resulted from the modulation of microglial metabolic reprogramming. We further showed that PFP-OLNDs regulated microglial metabolic reprogramming through the AKT-mTOR-HIF-1α pathway. Collectively, our findings suggest that the novel PFP-OLNDs constructed in this study alleviate microglia-mediated central inflammatory reactions through metabolic reprogramming.
Collapse
Affiliation(s)
- Wanxian Luo
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chuanhui Xu
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Linxi Li
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yunxiang Ji
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yezhong Wang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yingjia Li
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yongyi Ye
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Massaro Cenere M, Tiberi M, Paldino E, D'Addario SL, Federici M, Giacomet C, Cutuli D, Matteocci A, Cossa F, Zarrilli B, Casadei N, Ledonne A, Petrosini L, Berretta N, Fusco FR, Chiurchiù V, Mercuri NB. Systemic inflammation accelerates neurodegeneration in a rat model of Parkinson's disease overexpressing human alpha synuclein. NPJ Parkinsons Dis 2024; 10:213. [PMID: 39500895 PMCID: PMC11538257 DOI: 10.1038/s41531-024-00824-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024] Open
Abstract
Increasing efforts have been made to elucidate how genetic and environmental factors interact in Parkinson's disease (PD). In the present study, we assessed the development of symptoms on a genetic PD rat model that overexpresses human α-synuclein (Snca+/+) at a presymptomatic age, exposed to a pro-inflammatory insult by intraperitoneal injection of lipopolysaccharide (LPS), using immunohistology, high-dimensional flow cytometry, constant potential amperometry, and behavioral analyses. A single injection of LPS into WT and Snca+/+ rats triggered long-lasting increase in the activation of pro-inflammatory microglial markers, monocytes, and T lymphocytes. However, only LPS Snca+/+ rats showed dopaminergic neuronal loss in the substantia nigra pars compacta (SNpc), associated with a reduction in the release of evoked dopamine in the striatum. No significant changes were observed in the behavioral domain. We propose our double-hit animal as a reliable model to investigate the mechanisms whereby α-synuclein and inflammation interact to promote neurodegeneration in PD.
Collapse
Affiliation(s)
- Mariangela Massaro Cenere
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy.
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Marta Tiberi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Laboratory of Resolution of Neuroinflammation, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Emanuela Paldino
- Laboratory of Neuroanatomy, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Sebastian Luca D'Addario
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Mauro Federici
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Cecilia Giacomet
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Debora Cutuli
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Alessandro Matteocci
- Laboratory of Resolution of Neuroinflammation, Santa Lucia Foundation IRCCS, Rome, Italy
- PhD program in Immunology, Molecular Medicine and Applied biotechnologies, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Francesca Cossa
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Beatrice Zarrilli
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura Petrosini
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Nicola Berretta
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | | | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, Santa Lucia Foundation IRCCS, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Nicola B Mercuri
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy.
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
3
|
da Silva AAF, Fiadeiro MB, Bernardino LI, Fonseca CSP, Baltazar GMF, Cristóvão ACB. "Lipopolysaccharide-induced animal models for neuroinflammation - An overview.". J Neuroimmunol 2024; 387:578273. [PMID: 38183948 DOI: 10.1016/j.jneuroim.2023.578273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 01/08/2024]
Abstract
Neuroinflammation is a pathological mechanism contributing to neurodegenerative diseases. For in-depth studies of neuroinflammation, several animal models reported reproducing behavioral dysfunctions and cellular pathological mechanisms induced by brain inflammation. One of the most popular models of neuroinflammation is the one generated by lipopolysaccharide exposure. Despite its importance, the reported results using this model show high heterogeneity, making it difficult to analyze and compare the outcomes between studies. Therefore, the current review aims to summarize the different experimental paradigms used to reproduce neuroinflammation by lipopolysaccharide exposure and its respective outcomes, helping to choose the model that better suits each specific research aim.
Collapse
Affiliation(s)
- Ana Alexandra Flores da Silva
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal; NeuroSoV/Fastprinciple-Lda, UBIMedical, Universidade da Beira Interior, Covilhã, Portugal
| | - Mariana Bernardo Fiadeiro
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal; NeuroSoV/Fastprinciple-Lda, UBIMedical, Universidade da Beira Interior, Covilhã, Portugal
| | | | | | | | - Ana Clara Braz Cristóvão
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal; NeuroSoV/Fastprinciple-Lda, UBIMedical, Universidade da Beira Interior, Covilhã, Portugal.
| |
Collapse
|
4
|
Chu CH, Chen JS, Chan YL, Lu WJ, Huang YT, Mao PC, Sze CI, Sun HS. TIAM2S-positive microglia enhance inflammation and neurotoxicity through soluble ICAM-1-mediated immune priming. FASEB J 2023; 37:e23242. [PMID: 37801065 DOI: 10.1096/fj.202300462rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 09/05/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
TIAM Rac1-associated GEF 2 short form (TIAM2S) as an oncoprotein alters the immunity of peripheral immune cells to construct an inflammatory tumor microenvironment. However, its role in the activation of microglia, the primary innate immune cells of the brain, and neuroinflammation remains unknown. This study investigated the mechanism underlying TIAM2S shapes immune properties of microglia to facilitate neuron damage. Human microglial clone 3 cell line (HMC3) and human brain samples were applied to determine the presence of TIAM2S in microglia by western blots and double immunostaining. Furthermore, TIAM2S transgenic mice combined with multiple reconstituted primary neuron-glial culture systems and a cytokine array were performed to explore how TIAM2S shaped immune priming of microglia and participated in lipopolysaccharide (LPS)-induced neuron damage. TIAM2S protein was detectable in HMC3 cells and presented in a small portion (~11.1%) of microglia in human brains referred to as TIAM2S-positive microglia. With the property of secreted soluble factor-mediated immune priming, TIAM2S-positive microglia enhanced LPS-induced neuroinflammation and neural damage in vivo and in vitro. The gain- and loss-of-function experiments showed soluble intercellular adhesion molecule-1 (sICAM-1) participated in neurotoxic immune priming of TIAM2S+ microglia. Together, this study demonstrated a novel TIAM2S-positive microglia subpopulation enhances inflammation and neurotoxicity through sICAM-1-mediated immune priming.
Collapse
Affiliation(s)
- Chun-Hsien Chu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jia-Shing Chen
- School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| | - Ya-Ling Chan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Jen Lu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Te Huang
- Department of Geriatrics and Gerontology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Pin-Cheng Mao
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-I Sze
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - H Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
5
|
Dovonou A, Bolduc C, Soto Linan V, Gora C, Peralta Iii MR, Lévesque M. Animal models of Parkinson's disease: bridging the gap between disease hallmarks and research questions. Transl Neurodegener 2023; 12:36. [PMID: 37468944 PMCID: PMC10354932 DOI: 10.1186/s40035-023-00368-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by motor and non-motor symptoms. More than 200 years after its first clinical description, PD remains a serious affliction that affects a growing proportion of the population. Prevailing treatments only alleviate symptoms; there is still neither a cure that targets the neurodegenerative processes nor therapies that modify the course of the disease. Over the past decades, several animal models have been developed to study PD. Although no model precisely recapitulates the pathology, they still provide valuable information that contributes to our understanding of the disease and the limitations of our treatment options. This review comprehensively summarizes the different animal models available for Parkinson's research, with a focus on those induced by drugs, neurotoxins, pesticides, genetic alterations, α-synuclein inoculation, and viral vector injections. We highlight their characteristics and ability to reproduce PD-like phenotypes. It is essential to realize that the strengths and weaknesses of each model and the induction technique at our disposal are determined by the research question being asked. Our review, therefore, seeks to better aid researchers by ensuring a concrete discernment of classical and novel animal models in PD research.
Collapse
Affiliation(s)
- Axelle Dovonou
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Cyril Bolduc
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Victoria Soto Linan
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Charles Gora
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Modesto R Peralta Iii
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Martin Lévesque
- CERVO Brain Research Centre, 2601, Chemin de la Canardière, Québec, QC, G1J 2G3, Canada.
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
6
|
Brown GC, Camacho M, Williams‐Gray CH. The Endotoxin Hypothesis of Parkinson's Disease. Mov Disord 2023; 38:1143-1155. [PMID: 37157885 PMCID: PMC10947365 DOI: 10.1002/mds.29432] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023] Open
Abstract
The endotoxin hypothesis of Parkinson's disease (PD) is the idea that lipopolysaccharide (LPS) endotoxins contribute to the pathogenesis of this disorder. LPS endotoxins are found in, and released from, the outer membrane of Gram-negative bacteria, for example in the gut. It is proposed that gut dysfunction in early PD leads to elevated LPS levels in the gut wall and blood, which promotes both α-synuclein aggregation in the enteric neurons and a peripheral inflammatory response. Communication to the brain via circulating LPS and cytokines in the blood and/or the gut-brain axis leads to neuroinflammation and spreading of α-synuclein pathology, exacerbating neurodegeneration in brainstem nuclei and loss of dopaminergic neurons in the substantia nigra, and manifesting in the clinical symptoms of PD. The evidence supporting this hypothesis includes: (1) gut dysfunction, permeability, and bacterial changes occur early in PD, (2) serum levels of LPS are increased in a proportion of PD patients, (3) LPS induces α-synuclein expression, aggregation, and neurotoxicity, (4) LPS causes activation of peripheral monocytes leading to inflammatory cytokine production, and (5) blood LPS causes brain inflammation and specific loss of midbrain dopaminergic neurons, mediated by microglia. If the hypothesis is correct, then treatment options might include: (1) changing the gut microbiome, (2) reducing gut permeability, (3) reducing circulating LPS levels, or (4) blocking the response of immune cells and microglia to LPS. However, the hypothesis has a number of limitations and requires further testing, in particular whether reducing LPS levels can reduce PD incidence, progression, or severity. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Guy C. Brown
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Marta Camacho
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
7
|
Martin M, Pusceddu MM, Teichenné J, Negra T, Connolly A, Escoté X, Torrell Galceran H, Cereto Massagué A, Samarra Mestre I, Del Pino Rius A, Romero-Gimenez J, Egea C, Alcaide-Hidalgo JM, Del Bas JM. Preventive Treatment with Astaxanthin Microencapsulated with Spirulina Powder, Administered in a Dose Range Equivalent to Human Consumption, Prevents LPS-Induced Cognitive Impairment in Rats. Nutrients 2023; 15:2854. [PMID: 37447181 DOI: 10.3390/nu15132854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Cognitive alterations are a common feature associated with many neurodegenerative diseases and are considered a major health concern worldwide. Cognitive alterations are triggered by microglia activation and oxidative/inflammatory processes in specific areas of the central nervous system. Consumption of bioactive compounds with antioxidative and anti-inflammatory effects, such as astaxanthin and spirulina, can help in preventing the development of these pathologies. In this study, we have investigated the potential beneficial neuroprotective effects of a low dose of astaxanthin (ASX) microencapsulated within spirulina (ASXSP) in female rats to prevent the cognitive deficits associated with the administration of LPS. Alterations in memory processing were evaluated in the Y-Maze and Morris Water Maze (MWM) paradigms. Changes in microglia activation and in gut microbiota content were also investigated. Our results demonstrate that LPS modified long-term memory in the MWM and increased microglia activation in the hippocampus and prefrontal cortex. Preventive treatment with ASXSP ameliorated LPS-cognitive alterations and microglia activation in both brain regions. Moreover, ASXSP was able to partially revert LPS-induced gut dysbiosis. Our results demonstrate the neuroprotective benefits of ASX when microencapsulated with spirulina acting through different mechanisms, including antioxidant, anti-inflammatory and, probably, prebiotic actions.
Collapse
Affiliation(s)
- Miquel Martin
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Matteo M Pusceddu
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Joan Teichenné
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | | | | | - Xavier Escoté
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Helena Torrell Galceran
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Adrià Cereto Massagué
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Iris Samarra Mestre
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Antoni Del Pino Rius
- Eurecat-Centre Tecnològic de Catalunya, Centre for Omic Sciences, Joint Unit Eurecat-Universitat Rovira i Virgili, Unique Scientific and Technical Infrastructure (ICTS), 43204 Reus, Spain
| | - Jordi Romero-Gimenez
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | - Cristina Egea
- Eurecat-Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| | | | - Josep Maria Del Bas
- Eurecat-Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain
| |
Collapse
|
8
|
D Magalhães J, Candeias E, Melo-Marques I, Silva DF, Esteves AR, Empadinhas N, Cardoso SM. Intestinal infection triggers mitochondria-mediated α-synuclein pathology: relevance to Parkinson's disease. Cell Mol Life Sci 2023; 80:166. [PMID: 37249642 PMCID: PMC11072242 DOI: 10.1007/s00018-023-04819-3] [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: 05/05/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 05/31/2023]
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disease characterized by the loss of dopaminergic neurons in the midbrain. In the prodromal phase several autonomic symptoms including orthostatic hypotension and constipation are correlated with increased α-synuclein pathology in peripheral tissues. It is currently accepted that some idiopathic PD cases may start in the gut (body-first PD) with accumulation of pathological α-synuclein in enteric neurons that may subsequently propagate caudo-rostrally to the central nervous system. In addition to the already-established regulation of synaptic vesicle trafficking, α-synuclein also seems to play a role in neuronal innate immunity after infection. Our goal was to understand if seeding the gut with the foodborne pathogen Listeria monocytogenes by oral gavage would impact gut immunity and eventually the central nervous system. Our results demonstrate that L. monocytogenes infection induced oligomerization of α-synuclein in the ileum, along with a pronounced pro-inflammatory local and systemic response that ultimately culminated in neuronal mitochondria dysfunction. We propose that, having evolved from ancestral endosymbiotic bacteria, mitochondria may be directly targeted by virulence factors of intracellular pathogens, and that mitochondrial dysfunction and fragmentation resulting also from the activation of the innate immune system at the gut level, trigger innate immune responses in midbrain neurons, which include α-synuclein oligomerization and neuroinflammation, all of which hallmarks of PD.
Collapse
Affiliation(s)
- João D Magalhães
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Emanuel Candeias
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Inês Melo-Marques
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Diana F Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - A Raquel Esteves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Nuno Empadinhas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
| | - Sandra Morais Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- Faculty of Medicine, Institute of Cellular and Molecular Biology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
9
|
Wang J, Miao X, Sun Y, Li S, Wu A, Wei C. Dopaminergic System in Promoting Recovery from General Anesthesia. Brain Sci 2023; 13:brainsci13040538. [PMID: 37190503 DOI: 10.3390/brainsci13040538] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 05/17/2023] Open
Abstract
Dopamine is an important neurotransmitter that plays a biological role by binding to dopamine receptors. The dopaminergic system regulates neural activities, such as reward and punishment, memory, motor control, emotion, and sleep-wake. Numerous studies have confirmed that the dopaminergic system has the function of maintaining wakefulness in the body. In recent years, there has been increasing evidence that the sleep-wake cycle in the brain has similar neurobrain network mechanisms to those associated with the loss and recovery of consciousness induced by general anesthesia. With the continuous development and innovation of neurobiological techniques, the dopaminergic system has now been proved to be involved in the emergence from general anesthesia through the modulation of neuronal activity. This article is an overview of the dopaminergic system and the research progress into its role in wakefulness and general anesthesia recovery. It provides a theoretical basis for interpreting the mechanisms regulating consciousness during general anesthesia.
Collapse
Affiliation(s)
- Jinxu Wang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaolei Miao
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yi Sun
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Sijie Li
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
10
|
Deng I, Bobrovskaya L. Lipopolysaccharide mouse models for Parkinson's disease research: a critical appraisal. Neural Regen Res 2022; 17:2413-2417. [PMID: 35535880 PMCID: PMC9120679 DOI: 10.4103/1673-5374.331866] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/20/2021] [Accepted: 11/30/2021] [Indexed: 12/25/2022] Open
Abstract
Parkinson's disease, the most common movement disorder, has a strong neuroinflammatory aspect. This is evident by increased pro-inflammatory cytokines in the serum, and the presence of activated microglial cells, and inflammatory cytokines in the substantia nigra of post-mortem brains as well as cerebrospinal fluid of Parkinson's disease patients. The central and peripheral neuroinflammatory aspects of Parkinson's disease can be investigated in vivo via administration of the inflammagen lipopolysaccharide, a component of the cell wall of gram-negative bacteria. In this mini-review, we will critically evaluate different routes of lipopolysaccharide administration (including intranasal systemic and stereotasic), their relevance to clinical Parkinson's disease as well as the recent findings in lipopolysaccharide mouse models. We will also share our own experiences with systemic and intrastriatal lipopolysaccharide models in C57BL/6 mice and will discuss the usefulness of lipopolysaccharide mouse models for future research in the field.
Collapse
Affiliation(s)
- Isaac Deng
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
11
|
Brandi E, Torres-Garcia L, Svanbergsson A, Haikal C, Liu D, Li W, Li JY. Brain region-specific microglial and astrocytic activation in response to systemic lipopolysaccharides exposure. Front Aging Neurosci 2022; 14:910988. [PMID: 36092814 PMCID: PMC9459169 DOI: 10.3389/fnagi.2022.910988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/26/2022] [Indexed: 11/24/2022] Open
Abstract
Microglia cells are the macrophage population within the central nervous system, which acts as the first line of the immune defense. These cells present a high level of heterogeneity among different brain regions regarding morphology, cell density, transcriptomes, and expression of different inflammatory mediators. This region-specific heterogeneity may lead to different neuroinflammatory responses, influencing the regional involvement in several neurodegenerative diseases. In this study, we aimed to evaluate microglial response in 16 brain regions. We compared different aspects of the microglial response, such as the extension of their morphological changes, sensitivity, and ability to convert an acute inflammatory response to a chronic one. Then, we investigated the synaptic alterations followed by acute and chronic inflammation in substantia nigra. Moreover, we estimated the effect of partial ablation of fractalkine CX3C receptor 1 (CX3CR1) on microglial response. In the end, we briefly investigated astrocytic heterogeneity and activation. To evaluate microglial response in different brain regions and under the same stimulus, we induced a systemic inflammatory reaction through a single intraperitoneal (i.p.) injection of lipopolysaccharides (LPS). We performed our study using C57BL6 and CX3CR1+/GFP mice to investigate microglial response in different regions and the impact of CX3CR1 partial ablation. We conducted a topographic study quantifying microglia alterations in 16 brain regions through immunohistochemical examination and computational image analysis. Assessing Iba1-immunopositive profiles and the density of the microglia cells, we have observed significant differences in region-specific responses of microglia populations in all parameters considered. Our results underline the peculiar microglial inflammation in the substantia nigra pars reticulata (SNpr). Here and in concomitance with the acute inflammatory response, we observed a transient decrease of dopaminergic dendrites and an alteration of the striato-nigral projections. Additionally, we found a significant decrease in microglia response and the absence of chronic inflammation in CX3CR1+/GFP mice compared to the wild-type ones, suggesting the CX3C axis as a possible pharmacological target against neuroinflammation induced by an increase of systemic tumor necrosis factor-alpha (TNFα) or/and LPS. Finally, we investigated astrocytic heterogeneity in this model. We observed different distribution and morphology of GFAP-positive astrocytes, a heterogeneous response under inflammatory conditions, and a decrease in their activation in CX3CR1 partially ablated mice compared with C57BL6 mice. Altogether, our data confirm that microglia and astrocytes heterogeneity lead to a region-specific inflammatory response in presence of a systemic TNFα or/and LPS treatment.
Collapse
Affiliation(s)
- Edoardo Brandi
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Laura Torres-Garcia
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Alexander Svanbergsson
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Caroline Haikal
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Di Liu
- Health Sciences Institute, China Medical University, Shenyang, China
| | - Wen Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Health Sciences Institute, China Medical University, Shenyang, China
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Health Sciences Institute, China Medical University, Shenyang, China
- *Correspondence: Jia-Yi Li, ,
| |
Collapse
|
12
|
Kim J, Lee HJ, Park JH, Cha BY, Hoe HS. Nilotinib modulates LPS-induced cognitive impairment and neuroinflammatory responses by regulating P38/STAT3 signaling. J Neuroinflammation 2022; 19:187. [PMID: 35841100 PMCID: PMC9288088 DOI: 10.1186/s12974-022-02549-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/05/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In chronic myelogenous leukemia, reciprocal translocation between chromosome 9 and chromosome 22 generates a chimeric protein, Bcr-Abl, that leads to hyperactivity of tyrosine kinase-linked signaling transduction. The therapeutic agent nilotinib inhibits Bcr-Abl/DDR1 and can cross the blood-brain barrier, but its potential impact on neuroinflammatory responses and cognitive function has not been studied in detail. METHODS The effects of nilotinib in vitro and in vivo were assessed by a combination of RT-PCR, real-time PCR, western blotting, ELISA, immunostaining, and/or subcellular fractionation. In the in vitro experiments, the effects of 200 ng/mL LPS or PBS on BV2 microglial cells, primary microglia or primary astrocytes pre- or post-treated with 5 µM nilotinib or vehicle were evaluated. The in vivo experiments involved wild-type mice administered a 7-day course of daily injections with 20 mg/kg nilotinib (i.p.) or vehicle before injection with 10 mg/kg LPS (i.p.) or PBS. RESULTS In BV2 microglial cells, pre- and post-treatment with nilotinib altered LPS-induced proinflammatory/anti-inflammatory cytokine mRNA levels by suppressing AKT/P38/SOD2 signaling. Nilotinib treatment also significantly downregulated LPS-stimulated proinflammatory cytokine levels in primary microglia and primary astrocytes by altering P38/STAT3 signaling. Experiments in wild-type mice showed that nilotinib administration affected LPS-mediated microglial/astroglial activation in a brain region-specific manner in vivo. In addition, nilotinib significantly reduced proinflammatory cytokine IL-1β, IL-6 and COX-2 levels and P38/STAT3 signaling in the brain in LPS-treated wild-type mice. Importantly, nilotinib treatment rescued LPS-mediated spatial working memory impairment and cortical dendritic spine number in wild-type mice. CONCLUSIONS Our results indicate that nilotinib can modulate neuroinflammatory responses and cognitive function in LPS-stimulated wild-type mice.
Collapse
Affiliation(s)
- Jieun Kim
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Korea
| | - Hyun-Ju Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Korea
| | - Jin-Hee Park
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Korea
| | - Byung-Yoon Cha
- PharmacoRex Co., Ltd., 20 Techno 1-ro, Yuseong-gu, Daejeon, 34016, Korea
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Korea. .,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Korea.
| |
Collapse
|
13
|
Somensi N, Lopes SC, Gasparotto J, Mayer Gonçalves R, Tiefensee-Ribeiro C, Oppermann Peixoto D, Ozorio Brum P, Pinho CM, Agnes JP, Santos L, de Oliveira J, Spiller F, Fonseca Moreira JC, Zanotto-Filho A, Prediger RD, Pens Gelain D. Role of toll-like receptor 4 and sex in 6-hydroxydopamine-induced behavioral impairments and neurodegeneration in mice. Neurochem Int 2021; 151:105215. [PMID: 34710535 DOI: 10.1016/j.neuint.2021.105215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/02/2021] [Accepted: 10/21/2021] [Indexed: 11/19/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive loss of the nigrostriatal dopaminergic neurons that are associated with motor alterations and non-motor manifestations (such as depression). Neuroinflammation is a process with a critical role in the pathogenesis of PD. In this regard, toll-like receptor 4 (TLR4) is a central mediator of immune response in PD. Moreover, there are gender-related differences in the incidence, prevalence, and clinical features of PD. Therefore, we aimed to elucidate the role of TLR4 in the sex-dependent response to dopaminergic denervation induced by 6-hydroxydopamine (6-OHDA) in mice. Female and male adult wildtype (WT) and TLR4 knockout (TLR4-/-) mice were administered with unilateral injection of 6-OHDA in the dorsal striatum, and non-motor and motor impairments were evaluated for 30 days, followed by biochemistry analysis in the substantia nigra pars compacta (SNc), dorsal striatum, and dorsoventral cortex. Early non-motor impairments (i.e., depressive-like behavior and spatial learning deficits) induced by 6-OHDA were observed in the male WT mice but not in male TLR4-/- or female mice. Motor alterations were observed after administration of 6-OHDA in both strains, and the lack of TLR4 was also related to motor commitment. Moreover, ablation of TLR4 prevented 6-OHDA-induced dopaminergic denervation and microgliosis in the SNc, selectively in female mice. These results reinforced the existence of sex-biased alterations in PD and indicated TLR4 as a promising therapeutic target for the motor and non-motor symptoms of PD, which will help counteract the neuroinflammatory and neurodegenerative processes.
Collapse
Affiliation(s)
- Nauana Somensi
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Samantha Cristiane Lopes
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Juciano Gasparotto
- Instituto de Ciências Biomédicas - Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700. CEP: 37130-001. Centro - Alfenas/MG, Alfenas, Minas Gerais, Brazil
| | - Rosângela Mayer Gonçalves
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Camila Tiefensee-Ribeiro
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daniel Oppermann Peixoto
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pedro Ozorio Brum
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Cibele Martins Pinho
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Jonathan Paulo Agnes
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Lucas Santos
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Jade de Oliveira
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernando Spiller
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - José Cláudio Fonseca Moreira
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alfeu Zanotto-Filho
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Rui Daniel Prediger
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Universitário, Florianópolis, SC, Brazil
| | - Daniel Pens Gelain
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
14
|
The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res 2021; 172:105840. [PMID: 34450312 DOI: 10.1016/j.phrs.2021.105840] [Citation(s) in RCA: 309] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
Emerging evidence indicates that the gut microbiota play a crucial role in the bidirectional communication between the gut and the brain suggesting that the gut microbes may shape neural development, modulate neurotransmission and affect behavior, and thereby contribute to the pathogenesis and/or progression of many neurodevelopmental, neuropsychiatric, and neurological conditions. This review summarizes recent data on the role of microbiota-gut-brain axis in the pathophysiology of neuropsychiatric and neurological disorders including depression, anxiety, schizophrenia, autism spectrum disorders, Parkinson's disease, migraine, and epilepsy. Also, the involvement of microbiota in gut disorders co-existing with neuropsychiatric conditions is highlighted. We discuss data from both in vivo preclinical experiments and clinical reports including: (1) studies in germ-free animals, (2) studies exploring the gut microbiota composition in animal models of diseases or in humans, (3) studies evaluating the effects of probiotic, prebiotic or antibiotic treatment as well as (4) the effects of fecal microbiota transplantation.
Collapse
|
15
|
Peripheral inflammation induces long-term changes in tyrosine hydroxylase activation in the substantia nigra. Neurochem Int 2021; 146:105022. [PMID: 33746005 DOI: 10.1016/j.neuint.2021.105022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/02/2021] [Accepted: 03/12/2021] [Indexed: 11/22/2022]
Abstract
Inflammation plays a role in neuropathology. We hypothesised that inflammation, induced by a single intraperitoneal injection of lipopolysaccharide (LPS), would induce long-term changes in the regulation of tyrosine hydroxylase (TH) in the rat midbrain. The level of 12 cytokines was initially analysed from one day to six months after LPS injection to confirm that peripheral inflammation led to neuroinflammatory changes in the midbrain. In the substantia nigra (SN), the levels of 8 of the 12 measured cytokines was significantly increased at one day. Granulocyte-macrophage colony-stimulating factor showed a threefold increased level at 6 months. The ventral tegmental area (VTA) showed a completely different pattern, with no increases in the levels of the 12 cytokines at one day and no changes beyond one week. TH activity was determined using a tritiated water release assay, TH protein and phosphorylation levels (Ser19, Ser31 and Ser40) were determined using western blotting. TH-specific activity in the SN was unchanged at one day but was substantially increased at one week and one month with no concomitant increase in TH phosphorylation. Substantial changes in TH activation without changes in TH phosphorylation have not previously been observed in the brain in response to a range of stressors. TH-specific activity was increased in the SN, and in the caudate putamen, at 6 months and was associated with increased TH phosphorylation at Ser19 and Ser40 at both locations. TH-specific activity in the VTA showed only a transient increase at day one associated with increased phosphorylation at Ser19 and Ser31 but thereafter showed no changes. This study shows that inflammation induced by LPS generated two distinct long-term changes in TH activity in the SN that are caused by different mechanisms, but there were no long-term changes in the adjacent VTA.
Collapse
|
16
|
Kim C, Beilina A, Smith N, Li Y, Kim M, Kumaran R, Kaganovich A, Mamais A, Adame A, Iba M, Kwon S, Lee WJ, Shin SJ, Rissman RA, You S, Lee SJ, Singleton AB, Cookson MR, Masliah E. LRRK2 mediates microglial neurotoxicity via NFATc2 in rodent models of synucleinopathies. Sci Transl Med 2020; 12:eaay0399. [PMID: 33055242 PMCID: PMC8100991 DOI: 10.1126/scitranslmed.aay0399] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/04/2019] [Accepted: 03/31/2020] [Indexed: 12/15/2022]
Abstract
Synucleinopathies are neurodegenerative disorders characterized by abnormal α-synuclein deposition that include Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. The pathology of these conditions also includes neuronal loss and neuroinflammation. Neuron-released α-synuclein has been shown to induce neurotoxic, proinflammatory microglial responses through Toll-like receptor 2, but the molecular mechanisms involved are poorly understood. Here, we show that leucine-rich repeat kinase 2 (LRRK2) plays a critical role in the activation of microglia by extracellular α-synuclein. Exposure to α-synuclein was found to enhance LRRK2 phosphorylation and activity in mouse primary microglia. Furthermore, genetic and pharmacological inhibition of LRRK2 markedly diminished α-synuclein-mediated microglial neurotoxicity via lowering of tumor necrosis factor-α and interleukin-6 expression in mouse cultures. We determined that LRRK2 promoted a neuroinflammatory cascade by selectively phosphorylating and inducing nuclear translocation of the immune transcription factor nuclear factor of activated T cells, cytoplasmic 2 (NFATc2). NFATc2 activation was seen in patients with synucleinopathies and in a mouse model of synucleinopathy, where administration of an LRRK2 pharmacological inhibitor restored motor behavioral deficits. Our results suggest that modulation of LRRK2 and its downstream signaling mediator NFATc2 might be therapeutic targets for treating synucleinopathies.
Collapse
Affiliation(s)
- Changyoun Kim
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Alexandria Beilina
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nathan Smith
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Biochemistry and Redox Biology Center, University of Nebraska, Lincoln, NE 68588, USA
| | - Yan Li
- Protein/Peptide Sequencing Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Minhyung Kim
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ravindran Kumaran
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alice Kaganovich
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adamantios Mamais
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anthony Adame
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michiyo Iba
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Somin Kwon
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Won-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, and Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Soo-Jean Shin
- Department of Biomedical Sciences, Neuroscience Research Institute, and Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Robert A Rissman
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sungyong You
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, and Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Andrew B Singleton
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark R Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eliezer Masliah
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
17
|
Gentile F, Doneddu PE, Riva N, Nobile-Orazio E, Quattrini A. Diet, Microbiota and Brain Health: Unraveling the Network Intersecting Metabolism and Neurodegeneration. Int J Mol Sci 2020; 21:E7471. [PMID: 33050475 PMCID: PMC7590163 DOI: 10.3390/ijms21207471] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence gives support for the idea that extra-neuronal factors may affect brain physiology and its predisposition to neurodegenerative diseases. Epidemiological and experimental studies show that nutrition and metabolic disorders such as obesity and type 2 diabetes increase the risk of Alzheimer's and Parkinson's diseases after midlife, while the relationship with amyotrophic lateral sclerosis is uncertain, but suggests a protective effect of features of metabolic syndrome. The microbiota has recently emerged as a novel factor engaging strong interactions with neurons and glia, deeply affecting their function and behavior in these diseases. In particular, recent evidence suggested that gut microbes are involved in the seeding of prion-like proteins and their spreading to the central nervous system. Here, we present a comprehensive review of the impact of metabolism, diet and microbiota in neurodegeneration, by affecting simultaneously several aspects of health regarding energy metabolism, immune system and neuronal function. Advancing technologies may allow researchers in the future to improve investigations in these fields, allowing the buildup of population-based preventive interventions and development of targeted therapeutics to halt progressive neurologic disability.
Collapse
Affiliation(s)
- Francesco Gentile
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.G.); (N.R.)
- Neuromuscular and Neuroimmunology Service, Humanitas Clinical and Research Institute IRCCS, 20089 Milan, Italy; (P.E.D.); (E.N.-O.)
| | - Pietro Emiliano Doneddu
- Neuromuscular and Neuroimmunology Service, Humanitas Clinical and Research Institute IRCCS, 20089 Milan, Italy; (P.E.D.); (E.N.-O.)
| | - Nilo Riva
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.G.); (N.R.)
- Department of Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Eduardo Nobile-Orazio
- Neuromuscular and Neuroimmunology Service, Humanitas Clinical and Research Institute IRCCS, 20089 Milan, Italy; (P.E.D.); (E.N.-O.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
| | - Angelo Quattrini
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy; (F.G.); (N.R.)
| |
Collapse
|
18
|
Justin A, Ashwini P, Jose JA, Jeyarani V, Dhanabal SP, Manisha C, Mandal SP, Bhavimani G, Prabitha P, Yuvaraj S, Prashantha Kumar BR. Two Rationally Identified Novel Glitazones Reversed the Behavioral Dysfunctions and Exhibited Neuroprotection Through Ameliorating Brain Cytokines and Oxy-Radicals in ICV-LPS Neuroinflammatory Rat Model. Front Neurosci 2020; 14:530148. [PMID: 33100954 PMCID: PMC7546828 DOI: 10.3389/fnins.2020.530148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
The present study has planned to evaluate the neuroprotective activity of two novel glitazones in a neuroinflammatory rat model. Two novel glitazones were selected from an in-house virtual library of glitazones based on their docking scores against peroxisome proliferator-activated receptor-gamma (PPAR-γ) protein and other parameters studied in in silico computational studies. Initially, an acute oral toxicity study was carried out for glitazones in rats to assess the toxicity profile and to determine the therapeutic range for neuroprotective evaluation. Prior to induction of neuroinflammation, the treatments with glitazones (G1 and G2) and standard pioglitazone were made for four consecutive days to respective groups. On the fifth day, the neuroinflammation was induced by intracerebroventricular (ICV) administration of lipopolysaccharides (LPS) (2 μg/μl) using stereotaxic apparatus. After 7 days, the rats were subjected to behavioral assessment followed by neurobiochemical evaluation and histopathological studies. The pre-treatment with glitazones at two dose levels (15 and 30 mg/kg) has significantly reversed behavioral dysfunctions. Glitazones have shown significant reduction in the levels of LPO, NO, TNF-α, and IL-1β and also increased the levels of antioxidant enzymes such as SOD, CAT, and GSH in the brain of LPS-administered rats. The neuroprotection exhibited by two novel glitazones is comparable with standard pioglitazone. The PPAR-γ-dependent amelioration of cytokines and oxy-radicals released by novel glitazones during neuroinflammatory conditions may be attributed to the reversal of behavioral dysfunctions through preventing the degeneration of neurons in major regions of the brain.
Collapse
Affiliation(s)
- Antony Justin
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, India
| | - Premkumar Ashwini
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, India
| | - Jincy A Jose
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, India
| | - Victoria Jeyarani
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, India
| | - S P Dhanabal
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, India
| | - Chennu Manisha
- Department of Pharmacology, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Ooty, India
| | - Subhankar P Mandal
- Department of Pharmaceutical Chemistry, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Mysuru, India
| | - Guru Bhavimani
- Department of Pharmaceutical Chemistry, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Mysuru, India
| | - P Prabitha
- Department of Pharmaceutical Chemistry, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Mysuru, India
| | - S Yuvaraj
- Department of Pharmaceutical Chemistry, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Mysuru, India
| | - B R Prashantha Kumar
- Department of Pharmaceutical Chemistry, JSS Academy of Higher Education & Research, JSS College of Pharmacy, Mysuru, India
| |
Collapse
|
19
|
Wang Q, Oyarzabal EA, Song S, Wilson B, Santos JH, Hong JS. Locus coeruleus neurons are most sensitive to chronic neuroinflammation-induced neurodegeneration. Brain Behav Immun 2020; 87:359-368. [PMID: 31923552 PMCID: PMC7316605 DOI: 10.1016/j.bbi.2020.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/23/2019] [Accepted: 01/05/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) develops over decades through spatiotemporal stages that ascend from the brainstem to the forebrain. The mechanism behind this caudo-rostral neurodegeneration remains largely undefined. In unraveling this phenomenon, we recently developed a lipopolysaccharide (LPS)-elicited chronic neuroinflammatory mouse model that displays sequential losses of neurons in brainstem, substantia nigra, hippocampus and cortex. In this study, we aimed to investigate the mechanisms of caudo-rostral neurodegeneration and focused our efforts on the earliest neurodegeneration of vulnerable noradrenergic locus coeruleus (NE-LC) neurons in the brainstem. We found that compared with neurons in other brain regions, NE-LC neurons in untreated mice displayed high levels of mitochondrial oxidative stress that was severely exacerbated in the presence of LPS-elicited chronic neuroinflammation. In agreement, NE-LC neurons in LPS-treated mice displayed early reduction of complex IV expression and mitochondrial swelling and loss of cristae. Mechanistically, the activation of the superoxide-generating enzyme NADPH oxidase (NOX2) on NE-LC neurons was essential for their heightened vulnerability during chronic neuroinflammation. LPS induced early and high expressions of NOX2 in NE-LC neurons. Genetic or pharmacological inactivation of NOX2 markedly reduced mitochondrial oxidative stress and dysfunction in LPS-treated mice. Furthermore, inhibition of NOX2 significantly ameliorated LPS-induced NE-LC neurodegeneration. More importantly, post-treatment with NOX2 inhibitor diphenyleneiodonium when NE-LC neurodegeneration had already begun, still showed high efficacy in protecting NE-LC neurons from degeneration in LPS-treated mice. This study strongly supports that chronic neuroinflammation and NOX2 expression among vulnerable neuronal populations contribute to caudo-rostral degeneration in PD.
Collapse
Affiliation(s)
- Qingshan Wang
- School of Public Health, Dalian Medical University, Dalian, Liaoning, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China.
| | - Esteban A. Oyarzabal
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Sheng Song
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Belinda Wilson
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Janine H. Santos
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| |
Collapse
|
20
|
Harsanyiova J, Buday T, Kralova Trancikova A. Parkinson's Disease and the Gut: Future Perspectives for Early Diagnosis. Front Neurosci 2020; 14:626. [PMID: 32625058 PMCID: PMC7313629 DOI: 10.3389/fnins.2020.00626] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive degeneration of dopaminergic neurons, and at the cellular level by the formation of Lewy bodies in the central nervous system (CNS). However, the onset of the disease is believed to be localized to peripheral organs, particularly the gastrointestinal tract (GIT) and the olfactory bulb sooner before neuropathological changes occur in the CNS. Patients already in the pre-motor stage of PD suffer from various digestive problems and/or due to significant changes in the composition of the intestinal microbiome in this early stage of the disease. Detailed analyses of patient biopsies and autopsies as well as animal models of neuropathological changes characteristic of PD provided important information on the pathology or treatment of PD symptoms. However, presently is not clarified (i) the specific tissue in the GIT where the pathological processes associated with PD is initiated; (ii) the mechanism by which these processes are disseminated to the CNS or other tissues within the GIT; and (iii) which neuropathological changes could also serve as a reliable diagnostic marker of the premotor stages of PD, or (iv) which type of GIT tissue would be the most appropriate choice for routine examination of patient biopsies.
Collapse
Affiliation(s)
- Jana Harsanyiova
- Departmet of Pahophysiology, Jessenius Faculty of Medicine in Martin, Comenius University, Bratislava, Slovakia
| | - Tomas Buday
- Departmet of Pahophysiology, Jessenius Faculty of Medicine in Martin, Comenius University, Bratislava, Slovakia
| | - Alzbeta Kralova Trancikova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University, Bratislava, Slovakia
| |
Collapse
|
21
|
Vegeto E, Villa A, Della Torre S, Crippa V, Rusmini P, Cristofani R, Galbiati M, Maggi A, Poletti A. The Role of Sex and Sex Hormones in Neurodegenerative Diseases. Endocr Rev 2020; 41:5572525. [PMID: 31544208 PMCID: PMC7156855 DOI: 10.1210/endrev/bnz005] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDs) are a wide class of disorders of the central nervous system (CNS) with unknown etiology. Several factors were hypothesized to be involved in the pathogenesis of these diseases, including genetic and environmental factors. Many of these diseases show a sex prevalence and sex steroids were shown to have a role in the progression of specific forms of neurodegeneration. Estrogens were reported to be neuroprotective through their action on cognate nuclear and membrane receptors, while adverse effects of male hormones have been described on neuronal cells, although some data also suggest neuroprotective activities. The response of the CNS to sex steroids is a complex and integrated process that depends on (i) the type and amount of the cognate steroid receptor and (ii) the target cell type-either neurons, glia, or microglia. Moreover, the levels of sex steroids in the CNS fluctuate due to gonadal activities and to local metabolism and synthesis. Importantly, biochemical processes involved in the pathogenesis of NDs are increasingly being recognized as different between the two sexes and as influenced by sex steroids. The aim of this review is to present current state-of-the-art understanding on the potential role of sex steroids and their receptors on the onset and progression of major neurodegenerative disorders, namely, Alzheimer's disease, Parkinson's diseases, amyotrophic lateral sclerosis, and the peculiar motoneuron disease spinal and bulbar muscular atrophy, in which hormonal therapy is potentially useful as disease modifier.
Collapse
Affiliation(s)
- Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze della Salute (DiSS), Università degli Studi di Milano, Italy
| | - Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Valeria Crippa
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Paola Rusmini
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Riccardo Cristofani
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Mariarita Galbiati
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Scienze Farmaceutiche (DiSFarm), Università degli Studi di Milano, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Italy.,Dipartimento di Eccellenza di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Italy
| |
Collapse
|
22
|
Deng I, Corrigan F, Zhai G, Zhou XF, Bobrovskaya L. Lipopolysaccharide animal models of Parkinson's disease: Recent progress and relevance to clinical disease. Brain Behav Immun Health 2020; 4:100060. [PMID: 34589845 PMCID: PMC8474547 DOI: 10.1016/j.bbih.2020.100060] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative movement disorders which is characterised neuropathologically by progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the presence of Lewy bodies (made predominately of α-synuclein) in the surviving neurons. Animal models of PD have improved our understanding of the disease and have played a critical role in the development of neuroprotective agents. Neuroinflammation has been strongly implicated in the pathogenesis of PD, and recent studies have used lipopolysaccharide (LPS), a component of gram-negative bacteria and a potent activator of microglia cells, to mimic the inflammatory events in clinical PD. Modulating the inflammatory response could ameliorate PD associated complications and thus, it is essential to understand the extent to which LPS models reflect human PD. This review will outline the routes of administration of LPS such as stereotaxic, systemic and intranasal, their ability to recapitulate neuropathological markers of PD, and mechanisms of LPS induced toxicity. We will also discuss the ability of the models to replicate motor symptoms and non-motor symptoms of PD such as gastrointestinal dysfunction, olfactory dysfunction, anxiety, depression and cognitive dysfunction.
Collapse
Affiliation(s)
- Isaac Deng
- School of Pharmacy and Medical Sciences, Division of Health Sciences, Health and Biomedical Innovation Research Concentration, University of South Australia, Adelaide, South Australia, Australia
| | - Frances Corrigan
- School of Health Sciences, Division of Health Sciences, Health and Biomedical Innovation Research Concentration, University of South Australia, Adelaide, South Australia, Australia
| | - Guangxi Zhai
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan, 250012, China
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Division of Health Sciences, Health and Biomedical Innovation Research Concentration, University of South Australia, Adelaide, South Australia, Australia
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, Division of Health Sciences, Health and Biomedical Innovation Research Concentration, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
23
|
Zhao Z, Wang Y, Zhou R, Li Y, Gao Y, Tu D, Wilson B, Song S, Feng J, Hong JS, Yakel JL. A novel role of NLRP3-generated IL-1β in the acute-chronic transition of peripheral lipopolysaccharide-elicited neuroinflammation: implications for sepsis-associated neurodegeneration. J Neuroinflammation 2020; 17:64. [PMID: 32070376 PMCID: PMC7029475 DOI: 10.1186/s12974-020-1728-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Sepsis-associated acute brain inflammation, if unresolved, may cause chronic neuroinflammation and resultant neurodegenerative diseases. However, little is known how the transition from acute to chronic neuroinflammation, which is critical for the following progressive neurodegeneration, occurs in sepsis. The goal of this study was to investigate potential immune factors regulating the transition process using a widely used endotoxemia LPS mouse model. This model shows distinct acute and chronic phases of neuroinflammation and recapitulates many cardinal features of Parkinson's disease, thus, providing a unique opportunity for studying phase transition of neuroinflammation. METHODS C57BL/6 J, NLRP3-/-, and IL-1R1-/- mice were employed. Mild and severe endotoxemia were produced by LPS ip injection at 1 or 5 mg/kg. Neuroinflammation in vitro and in vivo was assessed with proinflammatory cytokine expression by qPCR or ELISA and microglial activation by immunohistochemical analysis. Neurodegeneration was measured by manual and stereological counts of nigral dopaminergic neurons and immunohistochemical analysis of protein nitrosylation and α-synuclein phosphorylation. RESULTS LPS-elicited initial increases in mouse brain mRNA levels of TNFα, IL-6, IL-1β, and MCP-1, and nigral microglial activation were not dose-related. By contrast, the delayed increase in brain mature IL-1β levels was dependent on LPS doses and protracted nigral microglial activation was only observed in high dose of LPS-treated mice. LPS-elicited increase in brain mature IL-1β but not IL-1α level was NLRP3-dependent. After high dose LPS treatment, deficiency of NLRP3 or IL-1R1 did not prevent the initiation of acute neuroinflammation but abolished chronic neuroinflammation. Genetic or pharmacological inhibition of the NLRP3-IL-1β axis repressed LPS-stimulated upregulation of chronic neuroinflammatory mediators including MHC-II, NOX2, and Mac1, and protected dopaminergic neurons. Ten months after LPS-elicited severe endotoxemia, nigral persisted microglial activation, elevated nitrosylated proteins and phosphorylated α-synuclein, and significant neuronal degeneration developed in wild-type mice but not in NLRP3-/- or IL-1R1-/- mice. CONCLUSIONS This study uncovers a novel role of the NLRP3-IL-1β signaling pathway in gauging the severity of sepsis-associated inflammation and determining whether acute neuroinflammation will resolve or transition to low grade chronic neuroinflammation. These findings also provide novel targets for developing therapy for severe systemic infection-related neurodegeneration.
Collapse
Affiliation(s)
- Zhan Zhao
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
- Institute of Infectious Diseases, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yubao Wang
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA.
- Institute of Infectious Diseases, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
| | - Ran Zhou
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Yi Li
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Yun Gao
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Dezhen Tu
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Belinda Wilson
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Sheng Song
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA.
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Jerrel L Yakel
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| |
Collapse
|
24
|
Tchessalova D, Tronson NC. Enduring and Sex-specific Changes in Hippocampal Gene Expression after a Subchronic Immune Challenge. Neuroscience 2020; 428:76-89. [PMID: 31917350 DOI: 10.1016/j.neuroscience.2019.12.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 01/14/2023]
Abstract
Major illnesses, including heart attack and sepsis, can cause cognitive impairments, depression, and progressive memory decline that persist long after recovery from the original illness. In rodent models of sepsis or subchronic immune challenge, memory deficits also persist for weeks or months, even in the absence of ongoing neuroimmune activation. This raises the question of what mechanisms in the brain mediate such persistent changes in neural function. Here, we used RNA-sequencing as a large-scale, unbiased approach to identify changes in hippocampal gene expression long after a subchronic immune challenge previously established to cause persistent memory impairments in both males and females. We observed enduring dysregulation of gene expression three months after the end of a subchronic immune challenge. Surprisingly, there were striking sex differences in both the magnitude of changes and the specific genes and pathways altered, where males showed persistent changes in both immune- and plasticity-related genes three months after immune challenge, whereas females showed few such changes. In contrast, females showed striking differential gene expression in response to a subsequent immune challenge. Thus, immune activation has enduring and sex-specific consequences for hippocampal gene expression and the transcriptional response to subsequent stimuli. Together with findings of long-lasting memory impairments after immune challenge, these data suggest that illnesses can cause enduring vulnerability to, cognitive decline, affective disorders, and memory impairments via dysregulation of transcriptional processes in the brain.
Collapse
Affiliation(s)
- Daria Tchessalova
- Department of Psychology and Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Natalie C Tronson
- Department of Psychology and Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
25
|
Gallardo-Orihuela A, Hervás-Corpión I, Hierro-Bujalance C, Sanchez-Sotano D, Jiménez-Gómez G, Mora-López F, Campos-Caro A, Garcia-Alloza M, Valor LM. Transcriptional correlates of the pathological phenotype in a Huntington's disease mouse model. Sci Rep 2019; 9:18696. [PMID: 31822756 PMCID: PMC6904489 DOI: 10.1038/s41598-019-55177-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023] Open
Abstract
Huntington disease (HD) is a fatal neurodegenerative disorder without a cure that is caused by an aberrant expansion of CAG repeats in exon 1 of the huntingtin (HTT) gene. Although a negative correlation between the number of CAG repeats and the age of disease onset is established, additional factors may contribute to the high heterogeneity of the complex manifestation of symptoms among patients. This variability is also observed in mouse models, even under controlled genetic and environmental conditions. To better understand this phenomenon, we analysed the R6/1 strain in search of potential correlates between pathological motor/cognitive phenotypical traits and transcriptional alterations. HD-related genes (e.g., Penk, Plk5, Itpka), despite being downregulated across the examined brain areas (the prefrontal cortex, striatum, hippocampus and cerebellum), exhibited tissue-specific correlations with particular phenotypical traits that were attributable to the contribution of the brain region to that trait (e.g., striatum and rotarod performance, cerebellum and feet clasping). Focusing on the striatum, we determined that the transcriptional dysregulation associated with HD was partially exacerbated in mice that showed poor overall phenotypical scores, especially in genes with relevant roles in striatal functioning (e.g., Pde10a, Drd1, Drd2, Ppp1r1b). However, we also observed transcripts associated with relatively better outcomes, such as Nfya (CCAAT-binding transcription factor NF-Y subunit A) plus others related to neuronal development, apoptosis and differentiation. In this study, we demonstrated that altered brain transcription can be related to the manifestation of HD-like symptoms in mouse models and that this can be extrapolated to the highly heterogeneous population of HD patients.
Collapse
Affiliation(s)
- Andrea Gallardo-Orihuela
- Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz, Spain.,Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - Irati Hervás-Corpión
- Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz, Spain.,Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - Carmen Hierro-Bujalance
- Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz, Spain.,Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Plaza Fragela, 11003, Cádiz, Spain
| | - Daniel Sanchez-Sotano
- Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz, Spain.,Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Plaza Fragela, 11003, Cádiz, Spain
| | - Gema Jiménez-Gómez
- Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz, Spain.,Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - Francisco Mora-López
- Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz, Spain.,Servicio de Inmunología, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - Antonio Campos-Caro
- Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz, Spain.,Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain
| | - Monica Garcia-Alloza
- Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz, Spain.,Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Plaza Fragela, 11003, Cádiz, Spain
| | - Luis M Valor
- Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz, Spain. .,Unidad de Investigación, Hospital Universitario Puerta del Mar, Av. Ana de Viya 21, 11009, Cádiz, Spain.
| |
Collapse
|
26
|
Jackson A, Forsyth CB, Shaikh M, Voigt RM, Engen PA, Ramirez V, Keshavarzian A. Diet in Parkinson's Disease: Critical Role for the Microbiome. Front Neurol 2019; 10:1245. [PMID: 31920905 PMCID: PMC6915094 DOI: 10.3389/fneur.2019.01245] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Parkinson's disease (PD) is the most common movement disorder affecting up to 1% of the population above the age of 60 and 4–5% of those above the age of 85. Little progress has been made on efforts to prevent disease development or halt disease progression. Diet has emerged as a potential factor that may prevent the development or slow the progression of PD. In this review, we discuss evidence for a role for the intestinal microbiome in PD and how diet-associated changes in the microbiome may be a viable approach to prevent or modify disease progression. Methods: We reviewed studies demonstrating that dietary components/foods were related to risk for PD. We reviewed evidence for the dysregulated intestinal microbiome in PD patients including abnormal shifts in the intestinal microbiota composition (i.e., dysbiosis) characterized by a loss of short chain fatty acid (SCFA) bacteria and increased lipopolysaccharide (LPS) bacteria. We also examined several candidate mechanisms by which the microbiota can influence PD including the NLRP3 inflammasome, insulin resistance, mitochondrial function, vagal nerve signaling. Results: The PD-associated microbiome is associated with decreased production of SCFA and increased LPS and it is believed that these changes may contribute to the development or exacerbation of PD. Diet robustly impacts the intestinal microbiome and the Western diet is associated with increased risk for PD whereas the Mediterranean diet (including high intake of dietary fiber) decreases PD risk. Mechanistically this may be the consequence of changes in the relative abundance of SCFA-producing or LPS-containing bacteria in the intestinal microbiome with effects on intestinal barrier function, endotoxemia (i.e., systemic LPS), NLRP3 inflammasome activation, insulin resistance, and mitochondrial dysfunction, and the production of factors such as glucagon like peptide 1 (GLP-1) and brain derived neurotrophic factor (BDNF) as well as intestinal gluconeogenesis. Conclusions: This review summarizes a model of microbiota-gut-brain-axis regulation of neuroinflammation in PD including several new mechanisms. We conclude with the need for clinical trials in PD patients to test this model for beneficial effects of Mediterranean based high fiber diets.
Collapse
Affiliation(s)
- Aeja Jackson
- Division of Digestive Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States.,Graduate College of Rush University, Chicago, IL, United States
| | - Christopher B Forsyth
- Division of Digestive Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States.,Graduate College of Rush University, Chicago, IL, United States
| | - Maliha Shaikh
- Division of Digestive Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Robin M Voigt
- Division of Digestive Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States.,Graduate College of Rush University, Chicago, IL, United States
| | - Phillip A Engen
- Division of Digestive Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Vivian Ramirez
- Division of Digestive Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States.,Graduate College of Rush University, Chicago, IL, United States
| | - Ali Keshavarzian
- Division of Digestive Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States.,Graduate College of Rush University, Chicago, IL, United States
| |
Collapse
|
27
|
Lipopolysaccharide-Induced Neuroinflammation as a Bridge to Understand Neurodegeneration. Int J Mol Sci 2019; 20:ijms20092293. [PMID: 31075861 PMCID: PMC6539529 DOI: 10.3390/ijms20092293] [Citation(s) in RCA: 314] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 12/19/2022] Open
Abstract
A large body of experimental evidence suggests that neuroinflammation is a key pathological event triggering and perpetuating the neurodegenerative process associated with many neurological diseases. Therefore, different stimuli, such as lipopolysaccharide (LPS), are used to model neuroinflammation associated with neurodegeneration. By acting at its receptors, LPS activates various intracellular molecules, which alter the expression of a plethora of inflammatory mediators. These factors, in turn, initiate or contribute to the development of neurodegenerative processes. Therefore, LPS is an important tool for the study of neuroinflammation associated with neurodegenerative diseases. However, the serotype, route of administration, and number of injections of this toxin induce varied pathological responses. Thus, here, we review the use of LPS in various models of neurodegeneration as well as discuss the neuroinflammatory mechanisms induced by this toxin that could underpin the pathological events linked to the neurodegenerative process.
Collapse
|
28
|
Tchessalova D, Tronson NC. Memory deficits in males and females long after subchronic immune challenge. Neurobiol Learn Mem 2019; 158:60-72. [PMID: 30611884 PMCID: PMC6879099 DOI: 10.1016/j.nlm.2019.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/30/2018] [Accepted: 01/02/2019] [Indexed: 01/18/2023]
Abstract
Memory impairments and cognitive decline persist long after recovery from major illness or injury, and correlate with increased risk of later dementia. Here we developed a subchronic peripheral immune challenge model to examine delayed and persistent memory impairments in females and in males. We show that intermittent injections of either lipopolysaccharides or Poly I:C cause memory decline in both sexes that are evident eight weeks after the immune challenge. Importantly, we observed sex-specific patterns of deficits. Females showed impairments in object recognition one week after challenge that persisted for at least eight weeks. In contrast, males had intact memory one week after the immune challenge but exhibited broad impairments in memory tasks including object recognition, and both context and tone fear conditioning several months later. The differential patterns of memory deficits in males and in females were observed without sustained microglial activation or changes in blood-brain barrier permeability. Together, these data suggest that transient neuroimmune activity results in differential vulnerabilities of females and males to memory decline after immune challenge. This model will be an important tool for determining the mechanisms in both sexes that contribute to memory impairments that develop over the weeks and months after recovery from illness. Future studies using this model will provide new insights into the role of chronic inflammation in the pathogenesis of long-lasting memory decline and dementias.
Collapse
Affiliation(s)
- Daria Tchessalova
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, United States; Department of Psychology, University of Michigan, Ann Arbor, United States.
| | - Natalie C Tronson
- Department of Psychology, University of Michigan, Ann Arbor, United States.
| |
Collapse
|
29
|
Checkoway H, Ilango S, Li W, Ray RM, Tanner CM, Hu SC, Wang X, Nielsen S, Gao DL, Thomas DB. Occupational exposures and parkinsonism among Shanghai women textile workers. Am J Ind Med 2018; 61:886-892. [PMID: 30198067 DOI: 10.1002/ajim.22906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2018] [Indexed: 11/09/2022]
Abstract
BACKGROUND Endotoxin, a contaminant of cotton dust, is an experimental model for parkinsonism (PS). METHODS We investigated associations between exposures to endotoxin, solvents, magnetic fields, and night shift work, and neurologist-determined PS among Shanghai women textile workers, including 537 retired cotton factory workers ages ≥50 years and an age-matched reference group of 286 retired textile workers not exposed to cotton dust. Repeat exams were conducted 2.5 years after enrollment among 467 cotton workers and 229 reference workers. RESULTS We identified 39 prevalent PS cases and 784 non-cases. No consistent or statistically significant associations were observed for endotoxin, solvents, magnetic fields, or shift work with PS risk, severity, or progression. CONCLUSIONS Despite the null findings, additional studies of endotoxin exposure and risk of PS in other well-characterized occupational cohorts are warranted in view of toxicological evidence that endotoxin is a pathogenic agent and its widespread occurrence in multiple industries worldwide.
Collapse
Affiliation(s)
- Harvey Checkoway
- Department of Family Medicine and Public Health; University of California; San Diego California
- Department of Neurosciences; University of California; San Diego California
| | - Sindana Ilango
- Department of Family Medicine and Public Health; University of California; San Diego California
- Graduate School of Public Health; San Diego State University; San Diego California
| | - Wenjin Li
- Public Health Sciences Division; Fred Hutchinson Cancer Research Center; Epidemiology Division; Seattle Washington
| | - Roberta M. Ray
- Public Health Sciences Division; Fred Hutchinson Cancer Research Center; Epidemiology Division; Seattle Washington
| | - Caroline M. Tanner
- Department of Neurology; University of California; San Francisco California
| | - Shu-Ching Hu
- Department of Neurology; University of Washington; Seattle Washington
| | - Xin Wang
- Department of Neurology; Zhong Shan Hospital; Fudan University; Shanghai PR China
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science; Fudan University; Shanghai China
| | - Susan Nielsen
- Department of Neurology; Washington University St. Louis; St. Louis Missouri
| | - Dao L. Gao
- Department of Epidemiology; Fudan University; Zhong Shan Hospital; Shanghai PR China
| | - David B. Thomas
- Public Health Sciences Division; Fred Hutchinson Cancer Research Center; Epidemiology Division; Seattle Washington
- Department of Epidemiology; University of Washington; Seattle Washington
| |
Collapse
|
30
|
Low-Grade Inflammation Aggravates Rotenone Neurotoxicity and Disrupts Circadian Clock Gene Expression in Rats. Neurotox Res 2018; 35:421-431. [PMID: 30328585 DOI: 10.1007/s12640-018-9968-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/08/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023]
Abstract
A single injection of LPS produced low-grade neuroinflammation leading to Parkinson's disease (PD) in mice several months later. Whether such a phenomenon occurs in rats and whether such low-grade neuroinflammation would aggravate rotenone (ROT) neurotoxicity and disrupts circadian clock gene/protein expressions were examined in this study. Male rats were given two injections of LPS (2.5-7.5 mg/kg), and neuroinflammation and dopamine neuron loss were evident 3 months later. Seven months after a single LPS (5 mg/kg) injection, rats received low doses of ROT (0.5 mg/kg, sc, 5 times/week for 4 weeks) to examine low-grade neuroinflammation on ROT toxicity. LPS plus ROT produced more pronounced non-motor and motor dysfunctions than LPS or ROT alone in behavioral tests, and decreased mitochondrial complex 1 activity, together with aggravated neuroinflammation and neuron loss. The expressions of clock core genes brain and muscle Arnt-like protein-1 (Bmal1), locomotor output cycles kaput (Clock), and neuronal PAS domain protein-2 (Npas2) were decreased in LPS, ROT, and LPS plus ROT groups. The expressions of circadian feedback genes Periods (Per1 and Per2) were also decreased, but Cryptochromes (Cry1 and Cry2) were unaltered. The circadian clock target genes nuclear receptor Rev-Erbα (Nr1d1), and D-box-binding protein (Dbp) expressions were also decreased. Consistent with the transcript levels, circadian clock protein BMAL1, CLOCK, NR1D1, and DBP were also decreased. Thus, LPS-induced chronic low-grade neuroinflammation potentiated ROT neurotoxicity and disrupted circadian clock gene/protein expression, suggesting a role of disrupted circadian in PD development and progression. Graphical Abstract ᅟ.
Collapse
|
31
|
Dirr ER, Ekhator OR, Blackwood R, Holden JG, Masliah E, Schultheis PJ, Fleming SM. Exacerbation of sensorimotor dysfunction in mice deficient in Atp13a2 and overexpressing human wildtype alpha-synuclein. Behav Brain Res 2018; 343:41-49. [PMID: 29407413 PMCID: PMC5829010 DOI: 10.1016/j.bbr.2018.01.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 12/26/2022]
Abstract
Loss of function mutations in the gene ATP13A2 are associated with Kufor-Rakeb Syndrome and Neuronal Ceroid Lipofuscinosis, the former designated as an inherited form of Parkinson's disease (PD). The function of ATP13A2 is unclear but in vitro studies indicate it is a lysosomal protein and may interact with the presynaptic protein alpha-synuclein (aSyn) and certain heavy metals. Accumulation of aSyn is a major component of lewy bodies, the pathological hallmark of PD. Atp13a2-deficient (13a2) mice develop age-dependent sensorimotor deficits, and accumulation of insoluble aSyn in the brain. To better understand the interaction between ATP13A2 and aSyn, double mutant mice with loss of Atp13a2 function combined with overexpression of human wildtype aSyn were generated. Female and male wildtype (WT), 13a2, aSyn, and 13a2-aSyn mice were tested on a battery of sensorimotor tests including adhesive removal, challenging beam traversal, spontaneous activity, gait, locomotor activity, and nest-building at 2, 4, and 6 months of age. Double mutant mice showed an earlier onset and accelerated alterations in sensorimotor function that were age, sex and test-dependent. Female 13a2-aSyn mice showed early and progressive dysfunction on the beam and in locomotor activity. In males, 13a2-aSyn mice showed more severe impairments in spontaneous activity and adhesive removal. Sex differences were also observed in aSyn and 13a2-aSyn mice on the beam, cylinder, and adhesive removal tests. In other tasks, double mutant mice displayed deficits similar to aSyn mice. These results indicate loss of Atp13a2 function exacerbates the sensorimotor phenotype in aSyn mice in an age and sex-dependent manner.
Collapse
Affiliation(s)
- Emily R Dirr
- Department of Neurology, School of Medicine, University of Cincinnati, USA
| | - Osunde R Ekhator
- Department of Neurology, School of Medicine, University of Cincinnati, USA
| | - Rachel Blackwood
- Department of Neurology, School of Medicine, University of Cincinnati, USA
| | - John G Holden
- Department of Neurology, School of Medicine, University of Cincinnati, USA
| | | | | | - Sheila M Fleming
- Department of Pharmaceutical Sciences and Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, USA.
| |
Collapse
|
32
|
Hou L, Wang K, Zhang C, Sun F, Che Y, Zhao X, Zhang D, Li H, Wang Q. Complement receptor 3 mediates NADPH oxidase activation and dopaminergic neurodegeneration through a Src-Erk-dependent pathway. Redox Biol 2018; 14:250-260. [PMID: 28978491 PMCID: PMC5975223 DOI: 10.1016/j.redox.2017.09.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/17/2017] [Accepted: 09/24/2017] [Indexed: 01/08/2023] Open
Abstract
Microglial NADPH oxidase (Nox2) plays a key role in chronic neuroinflammation and related dopaminergic neurodegeneration in Parkinson's disease (PD). However, the mechanisms behind Nox2 activation remain unclear. Here, we revealed the critical role of complement receptor 3 (CR3), a microglia-specific pattern recognition receptor, in Nox2 activation and subsequent dopaminergic neurodegeneration by using paraquat and maneb-induced PD model. Suppression or genetic deletion of CR3 impeded paraquat and maneb-induced activation of microglial Nox2, which was associated with attenuation of dopaminergic neurodegeneration. Mechanistic inquiry revealed that blocking CR3 reduced paraquat and maneb-induced membrane translocation of Nox2 cytosolic subunit p47phox, an essential step for Nox2 activation. Src and Erk (extracellular regulated protein kinases) were subsequently recognized as the downstream signals of CR3. Moreover, inhibition of Src or Erk impaired Nox2 activation in response to paraquat and maneb co-exposure. Finally, we found that CR3-deficient mice were more resistant to paraquat and maneb-induced Nox2 activation and nigral dopaminergic neurodegeneration as well as motor dysfunction than the wild type controls. Taken together, our results showed that CR3 regulated Nox2 activation and dopaminergic neurodegeneration through a Src-Erk-dependent pathway in a two pesticide-induced PD model, providing novel insights into the immune pathogenesis of PD.
Collapse
Affiliation(s)
- Liyan Hou
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Ke Wang
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Cong Zhang
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Fuqiang Sun
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Yuning Che
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China
| | - Xiulan Zhao
- Institute of Toxicology, School of Public Health, Shandong University, 44 West Wenhua Road, Jinan, Shandong, China
| | - Dan Zhang
- State Key Laboratory of Natural Products and Functions, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Huihua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, No. 9W. Lvshun South Road, Dalian 116044, China.
| |
Collapse
|
33
|
Hwang DS, Gu PS, Kim N, Jang YP, Oh MS. Effects of Rhei Undulati Rhizoma on lipopolysaccharide-induced neuroinflammation in vitro and in vivo. ENVIRONMENTAL TOXICOLOGY 2018; 33:23-31. [PMID: 28984087 DOI: 10.1002/tox.22463] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 06/07/2023]
Abstract
Neuroinflammation plays a critical role in the pathogenesis of degenerative brain diseases such as Alzheimer's disease and Parkinson's disease. Microglia are the major components of the brain immune system that regulate inflammatory processes. Activated microglia release pro-inflammatory factors and cytokines, resulting in neuronal cell death. We focused on inhibiting the activation of microglia from a stimulus as a strategy to search for neuroprotective drugs. Rhei Undulati Rhizoma (RUR) is traditionally used to treat various inflammatory disorders. In this study, we investigated whether RUR modulates inflammatory processes in lipopolysaccharide (LPS)-stimulated BV2 microglia cells and the mouse brain. RUR exerted anti-neuroinflammatory effects by inhibiting the production of nitric oxide and reactive oxygen species induced by LPS via the downregulation of transcription factors such as inducible nitric oxide synthase and cyclooxygenase-2 (COX-2) without causing cytotoxicity. RUR also regulated mitogen-activated protein kinase pathway by inhibiting phosphorylation of p38 and c-Jun N-terminal kinases and translocation of nuclear factor kappa B. Moreover, RUR attenuated LPS-induced glial activation and COX-2 expression in the substantia nigra and hippocampus of the mouse brain. These results indicate that RUR is a potential candidate to treat neurodegenerative diseases by regulating neuroinflammation.
Collapse
Affiliation(s)
- Deok-Sang Hwang
- Department of Oriental Gynecology, College of Oriental Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Pil Sung Gu
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Namkwon Kim
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Young Pyo Jang
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate school, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
34
|
Zhou X, Spittau B. Lipopolysaccharide-Induced Microglia Activation Promotes the Survival of Midbrain Dopaminergic Neurons In Vitro. Neurotox Res 2017; 33:856-867. [DOI: 10.1007/s12640-017-9842-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022]
|
35
|
Erro R, Brigo F, Tamburin S, Zamboni M, Antonini A, Tinazzi M. Nutritional habits, risk, and progression of Parkinson disease. J Neurol 2017; 265:12-23. [PMID: 29018983 DOI: 10.1007/s00415-017-8639-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022]
Abstract
Parkinson disease (PD) is a multifactorial disease, where a genetic predisposition combines with putative environmental risk factors. Mounting evidence suggests that the initial PD pathological manifestations may be located in the gut to subsequently affect brain areas. Moreover, several lines of research demonstrated that there are bidirectional connections between the central nervous system and the gut, the "gut-brain axis" that influences both brain and gastrointestinal function. This opens a potential therapeutic window suggesting that specific dietary strategies may interact with the disease process and influence the risk of PD or modify its course. Dietary components can also theoretically modulate the chronic activation of the inflammatory response that is associated with aging, the strongest risk factor for PD, that has been suggested to hasten the underlying neurodegenerative process in PD. Here, we reviewed the evidence supporting an association between certain dietary compound and either the risk or progression of PD and have provided an overview of the possible pathomechanisms linking nutrition and neurodegeneration. The results of our review would not support a clear role for any dietary components in reducing the risk or progression of PD. However, the evidence favouring a connection between gut abnormalities, inflammation, and neurodegeneration in PD have become too compelling to be ignored, so that further research, also in the field of nutritional genomics, is highly warranted.
Collapse
Affiliation(s)
- Roberto Erro
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Piazzale L.A. Scuro 10, 37134, Verona, Italy. .,Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy.
| | - Francesco Brigo
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Piazzale L.A. Scuro 10, 37134, Verona, Italy.,Department of Neurology, Franz Tappeiner Hospital, Merano, Italy
| | - Stefano Tamburin
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Piazzale L.A. Scuro 10, 37134, Verona, Italy
| | - Mauro Zamboni
- Section of Geriatrics, Department of Medicine, Division of Geriatrics, University of Verona, Verona, Italy
| | - Angelo Antonini
- Parkinson Unit, IRCCS Hospital San Camillo and 1st Neurology Clinic, AO Universitaria Padua, Padua, Italy
| | - Michele Tinazzi
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Piazzale L.A. Scuro 10, 37134, Verona, Italy
| |
Collapse
|
36
|
Girard-Joyal O, Ismail N. Effect of LPS treatment on tyrosine hydroxylase expression and Parkinson-like behaviors. Horm Behav 2017; 89:1-12. [PMID: 28025041 DOI: 10.1016/j.yhbeh.2016.12.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 01/18/2023]
Abstract
Puberty is a critical period of development during which the brain undergoes reorganizing and remodeling. Exposure to stress during this period is thought to interfere with normal brain development and increase susceptibility to mental illnesses. In female mice, pubertal exposure to lipopolysaccharide (LPS), a bacterial endotoxin, has been shown to alter sexual, anxiety-like, and depression-like behaviors and cognition in an enduring manner. However, the mechanisms underlying these effects remain unknown. The present study examined age and sex difference in tyrosine hydroxylase (TH) expression and dopamine-dependent and Parkinson-like behaviors following LPS treatment. The results show that LPS treatment during adulthood causes an enduring increase in TH expression in many of the brain regions examined. In contrast, there is no change in TH expression following LPS treatment during puberty. However, pubertal LPS treatment induces enduring behavioral deficits in tests of Parkinson-like behaviors, more so in male than in female mice. These results suggest that the low levels of TH following exposure to pubertal immune challenge may predispose mice to Parkinson-like behavior. These findings add to our understanding of stress and immune responses during puberty and their impact on mental health later in life.
Collapse
Affiliation(s)
| | - Nafissa Ismail
- School of Psychology, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
37
|
Humbert-Claude M, Duc D, Dwir D, Thieren L, Sandström von Tobel J, Begka C, Legueux F, Velin D, Maillard MH, Do KQ, Monnet-Tschudi F, Tenenbaum L. Tollip, an early regulator of the acute inflammatory response in the substantia nigra. J Neuroinflammation 2016; 13:303. [PMID: 27927222 PMCID: PMC5142340 DOI: 10.1186/s12974-016-0766-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/18/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Tollip is a ubiquitously expressed protein, originally described as a modulator of the IL-1R/TLR-NF-κB signaling pathways. Although this property has been well characterized in peripheral cells, and despite some evidence of its expression in the central nervous system, the role of Tollip in neuroinflammation remains poorly understood. The present study sought to explore the implication of Tollip in inflammation in the substantia nigra pars compacta, the structure affected in Parkinson's disease. METHODS We first investigated Tollip distribution in the midbrain by immunohistochemistry. Then, we addressed TLR4-mediated response by intra-nigral injections of lipopolysaccharide (LPS), a TLR4 agonist, on inflammatory markers in Tollip knockout (KO) and wild-type (WT) mice. RESULTS We report an unexpectedly high Tollip immunostaining in dopaminergic neurons of the mice brain. Second, intra-nigral injection of LPS led to increased susceptibility to neuroinflammation in Tollip KO compared to Tollip WT mice. This was demonstrated by a significant increase of tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), and interferon gamma (IFN-γ) messenger RNA (mRNA) in the midbrain of Tollip KO mice upon LPS injection. Consistently, brain rAAV viral vector transduction with a nuclear factor kappa B (NF-κB)-inducible reporter gene confirmed increased NF-κB activation in Tollip KO mice. Lastly, Tollip KO mice displayed higher inducible NO synthase (iNOS) production, both at the messenger and protein level when compared to LPS-injected WT mice. Tollip deletion also aggravated LPS-induced oxidative and nitrosative damages, as indicated by an increase of 8-oxo-2'-deoxyguanosine and nitrotyrosine immunostaining, respectively. CONCLUSIONS Altogether, these findings highlight a critical role of Tollip in the early phase of TLR4-mediated neuroinflammation. As brain inflammation is known to contribute to Parkinson's disease, Tollip may be a potential target for neuroprotection.
Collapse
Affiliation(s)
- Marie Humbert-Claude
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - D. Duc
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - D. Dwir
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Prilly, Lausanne, Switzerland
| | - L. Thieren
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | | | - C. Begka
- Service of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | | | - D. Velin
- Service of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - M. H. Maillard
- Service of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - K. Q. Do
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Prilly, Lausanne, Switzerland
| | - F. Monnet-Tschudi
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - L. Tenenbaum
- Laboratory of Cellular and Molecular Neurotherapies, Center for Neuroscience Research, Department of Clinical Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
38
|
Abstract
Inflammatory activation of microglia is a hallmark of several disorders of the central nervous system. In addition to protecting the brain against inflammatory insults, microglia are neuroprotective and play a significant role in maintaining neuronal connectivity, but the prolongation of an inflammatory status may limit the beneficial functions of these immune cells. The finding that estrogen receptors are present in monocyte-derived cells and that estrogens prevent and control the inflammatory response raise the question of the role that this sex steroid plays in the manifestation and progression of pathologies that have a clear sex difference in prevalence, such as multiple sclerosis, Parkinson's disease, and Alzheimer's disease. The present review aims to provide a critical review of the current literature on the actions of estrogen in microglia and on the involvement of estrogen receptors in the manifestation of selected neurological disorders. This current understanding highlights a research area that should be expanded to identify appropriate replacement therapies to slow the progression of such diseases.
Collapse
Affiliation(s)
- Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Elisabetta Vegeto
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Angelo Poletti
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| |
Collapse
|
39
|
Knapp DJ, Harper KM, Whitman BA, Zimomra Z, Breese GR. Stress and Withdrawal from Chronic Ethanol Induce Selective Changes in Neuroimmune mRNAs in Differing Brain Sites. Brain Sci 2016; 6:brainsci6030025. [PMID: 27472367 PMCID: PMC5039454 DOI: 10.3390/brainsci6030025] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/10/2016] [Accepted: 07/20/2016] [Indexed: 12/12/2022] Open
Abstract
Stress is a strong risk factor in alcoholic relapse and may exert effects that mimic aspects of chronic alcohol exposure on neurobiological systems. With the neuroimmune system becoming a prominent focus in the study of the neurobiological consequences of stress, as well as chronic alcohol exposure proving to be a valuable focus in this regard, the present study sought to compare the effects of stress and chronic ethanol exposure on induction of components of the neuroimmune system. Rats were exposed to either 1 h exposure to a mild stressor (restraint) or exposure to withdrawal from 15 days of chronic alcohol exposure (i.e., withdrawal from chronic ethanol, WCE) and assessed for neuroimmune mRNAs in brain. Restraint stress alone elevated chemokine (C–C motif) ligand 2 (CCL2), interleukin-1-beta (IL-1β), tumor necrosis factor alpha (TNFα) and toll-like receptor 4 (TLR4) mRNAs in the cerebral cortex within 4 h with a return to a control level by 24 h. These increases were not accompanied by an increase in corresponding proteins. Withdrawal from WCE also elevated cytokines, but did so to varying degrees across different cytokines and brain regions. In the cortex, stress and WCE induced CCL2, TNFα, IL-1β, and TLR4 mRNAs. In the hypothalamus, only WCE induced cytokines (CCL2 and IL-1β) while in the hippocampus, WCE strongly induced CCL2 while stress and WCE induced IL-1β. In the amygdala, only WCE induced CCL2. Finally—based on the previously demonstrated role of corticotropin-releasing factor 1 (CRF1) receptor inhibition in blocking WCE-induced cytokine mRNAs—the CRF1 receptor antagonist CP154,526 was administered to a subgroup of stressed rats and found to be inactive against induction of CCL2, TNFα, or IL-1β mRNAs. These differential results suggest that stress and WCE manifest broad neuroimmune effects in brain depending on the cytokine and brain region, and that CRF inhibition may not be a relevant mechanism in non-alcohol exposed animals. Overall, these effects are complex in terms of their neuroimmune targets and neuroanatomical specificity. Further investigation of the differential distribution of cytokine induction across neuroanatomical regions, individual cell types (e.g., neuronal phenotypes and glia), severity of chronic alcohol exposure, as well as across differing stress types may prove useful in understanding differential mechanisms of induction and for targeting select systems for pharmacotherapeutic intervention in alcoholism.
Collapse
Affiliation(s)
- Darin J Knapp
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Department of Psychiatry, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - Kathryn M Harper
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - Buddy A Whitman
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - Zachary Zimomra
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - George R Breese
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Department of Psychiatry, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Department of Pharmacology, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| |
Collapse
|
40
|
Role of the IL-1 Pathway in Dopaminergic Neurodegeneration and Decreased Voluntary Movement. Mol Neurobiol 2016; 54:4486-4495. [PMID: 27356916 PMCID: PMC5509814 DOI: 10.1007/s12035-016-9988-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/14/2016] [Indexed: 12/26/2022]
Abstract
Interleukin-1 (IL-1), a proinflammatory cytokine synthesized and released by activated microglia, can cause dopaminergic neurodegeneration leading to Parkinson’s disease (PD). However, it is uncertain whether IL-1 can act directly, or by exacerbating the harmful actions of other brain insults. To ascertain the role of the IL-1 pathway on dopaminergic neurodegeneration and motor skills during aging, we compared mice with impaired [caspase-1 knockout (casp1−/−)] or overactivated IL-1 activity [IL-1 receptor antagonist knockout (IL-1ra−/−)] to wild-type (wt) mice at young and middle age. Their motor skills were evaluated by the open-field and rotarod tests, and quantification of their dopamine neurons and activated microglia within the substantia nigra were performed by immunohistochemistry. IL-1ra−/− mice showed an age-related decline in motor skills, a reduced number of dopamine neurons, and an increase in activated microglia when compared to wt or casp1−/− mice. Casp1−/− mice had similar changes in motor skills and dopamine neurons, but fewer activated microglia cells than wt mice. Our results suggest that the overactivated IL-1 pathway occurring in IL-1ra−/− mice in the absence of inflammatory interventions (e.g., intracerebral injections performed in animal models of PD) increased activated microglia, decreased the number of dopaminergic neurons, and reduced their motor skills. Decreased IL-1 activity in casp1−/− mice did not yield clear protective effects when compared with wt mice. In summary, in the absence of overt brain insults, chronic activation of the IL-1 pathway may promote pathological aspects of PD per se, but its impairment does not appear to yield advantages over wt mice.
Collapse
|
41
|
Jiang L, Wu X, Wang S, Chen SH, Zhou H, Wilson B, Jin CY, Lu RB, Xie K, Wang Q, Hong JS. Clozapine metabolites protect dopaminergic neurons through inhibition of microglial NADPH oxidase. J Neuroinflammation 2016; 13:110. [PMID: 27184631 PMCID: PMC4869380 DOI: 10.1186/s12974-016-0573-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/06/2016] [Indexed: 01/02/2023] Open
Abstract
Background Clozapine, an atypical antipsychotic medication, has been effectively used to treat refractory schizophrenia. However, the clinical usage of clozapine is limited due to a high incidence of neutropenia or agranulocytosis. We previously reported that clozapine protected dopaminergic neurons through inhibition of microglial activation. The purpose of this study was to explore the neuroprotective effects of clozapine metabolites clozapine N-oxide (CNO) and N-desmethylclozapine (NDC), as well as their propensity to cause neutropenia. Methods The primary midbrain neuron-glia culture was applied to detect the neuroprotective and anti-inflammatory effect of clozapine and its metabolites in lipopolysaccharide (LPS) and MPP+-induced toxicity. And the subsequent mechanism was demonstrated by gp91phox mutant cell cultures as well as microgliosis cell lines. In vivo, to confirm the neuroprotective effect of clozapine and CNO, we measured the dopaminergic neuronal loss and rotarod motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-generated mouse Parkinson’s disease (PD) model. The neutropenia or agranulocytosis of clozapine and its metabolites was illustrated by white blood cell count of the treated mice. Results We found that, in midbrain neuron-glia cultures, CNO and NDC were more potent than clozapine in protecting dopaminergic neurons against LPS and MPP+-induced toxicity. CNO and NDC-afforded neuroprotection was linked to inhibition of microglia-mediated neuroinflammation, as demonstrated by abolished neuroprotection in microglia-depleted cultures and their capacity of inhibiting LPS-induced release of proinflammatory factors from activated microglia. NADPH oxidase (NOX2) was subsequently recognized as the main target of CNO and NDC since genetic ablation of gp91phox, the catalytic subunit of NOX2, abolished their neuroprotective effects. CNO and NDC inhibited NOX2 activation through interfering with the membrane translocation of the NOX2 cytosolic subunit, p47phox. The neuroprotective effects of CNO were further verified in vivo as shown by attenuation of dopaminergic neurodegeneration, motor deficits, and reactive microgliosis in MPTP-generated mouse PD model. More importantly, unlike clozapine, CNO did not lower the white blood cell count. Conclusions Altogether, our results show that clozapine metabolites elicited neuroprotection through inactivation of microglia by inhibiting NOX2. The robust neuroprotective effects and lack of neutropenia suggest that clozapine metabolites may be promising candidates for potential therapy for neurodegenerative diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0573-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lulu Jiang
- Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA.,Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Xuefei Wu
- Department of Physiology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Shuo Wang
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Shih-Heng Chen
- Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Hui Zhou
- Department of Occupational and Environmental Health, School of Public Health, Peking University, Beijing, 100191, China
| | - Belinda Wilson
- Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Chun-Yang Jin
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Ru-Band Lu
- Department of Psychiatry, Institute of Behavioral Medicine, Institute of Allied Health Sciences and Addiction Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.,Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, 35035, Taiwan
| | - Keqin Xie
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China.
| | - Qingshan Wang
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, Dalian, Liaoning, 116044, China.
| | - Jau-Shyong Hong
- Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| |
Collapse
|
42
|
Abstract
RATIONALE Alcoholism is a primary, chronic relapsing disease of brain reward, motivation, memory, and related circuitry. It is characterized by an individual's continued drinking despite negative consequences related to alcohol use, which is exemplified by alcohol use leading to clinically significant impairment or distress. Chronic alcohol consumption increases the expression of innate immune signaling molecules (ISMs) in the brain that alter cognitive processes and promote alcohol drinking. OBJECTIVES Unraveling the mechanisms of alcohol-induced neuroimmune gene induction is complicated by positive loops of multiple cytokines and other signaling molecules that converge on nuclear factor kappa-light-chain-enhancer of activated B cells and activator protein-1 leading to induction of additional neuroimmune signaling molecules that amplify and expand the expression of ISMs. RESULTS Studies from our laboratory employing reverse transcription polymerase chain reaction (RT-PCR) to assess mRNA, immunohistochemistry and Western blot analysis to assess protein expression, and others suggest that ethanol increases brain neuroimmune gene and protein expression through two distinct mechanisms involving (1) systemic induction of innate immune molecules that are transported from blood to the brain and (2) the direct release of high-mobility group box 1 (HMGB1) from neurons in the brain. Released HMGB1 signals through multiple receptors, particularly Toll-like receptor (TLR) 4, that potentiate cytokine receptor responses leading to a hyperexcitable state that disrupts neuronal networks and increases excitotoxic neuronal death. Innate immune gene activation in brain is persistent, consistent with the chronic relapsing disease that is alcoholism. Expression of HMGB1, TLRs, and other ISMs is increased several-fold in the human orbital frontal cortex, and expression of these molecules is highly correlated with each other as well as lifetime alcohol consumption and age of drinking onset. CONCLUSIONS The persistent and cumulative nature of alcohol on HMGB1 and TLR gene induction support their involvement in alcohol-induced long-term changes in brain function and neurodegeneration.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, CB# 7178, 1021 Thurston-Bowles Building, Chapel Hill, NC, 27599-7178, USA.
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, CB# 7178, 1021 Thurston-Bowles Building, Chapel Hill, NC, 27599-7178, USA
| |
Collapse
|
43
|
Kim C, Ojo-Amaize E, Spencer B, Rockenstein E, Mante M, Desplats P, Wrasidlo W, Adame A, Nchekwube E, Oyemade O, Okogun J, Chan M, Cottam H, Masliah E. Hypoestoxide reduces neuroinflammation and α-synuclein accumulation in a mouse model of Parkinson's disease. J Neuroinflammation 2015; 12:236. [PMID: 26683203 PMCID: PMC4683943 DOI: 10.1186/s12974-015-0455-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/11/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Deposition of α-synuclein and neuroinflammation are key pathological features of Parkinson's disease (PD). There is no cure for the disease; however, targeting the pathological features might be available to modulate the disease onset and progression. Hypoestoxide (HE) has been demonstrated as a NF-κB modulator, thereby acting as a potential anti-inflammatory and anti-cancer drug. METHODS In order to assess the effect of HE in a mouse model of PD, mThy1-α-syn transgenic mice received intraperitoneal (IP) injections of either vehicle or HE (5 mg/kg) daily for 4 weeks. RESULTS Treatment of HE decreased microgliosis, astrogliosis, and pro-inflammatory cytokine gene expression in α-syn transgenic mice. HE administration also prevented the loss of dopaminergic neurons and ameliorated motor behavioral deficits in the α-syn transgenic mice, and α-synuclein pathology was significantly reduced by treatment of HE. In addition, increased levels of nuclear phosphorylated NF-κB in the frontal cortex of α-syn transgenic mice were significantly reduced by HE administration. CONCLUSIONS These results support the therapeutic potential of HE for PD and other α-synuclein-related diseases.
Collapse
Affiliation(s)
- Changyoun Kim
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, 92093, USA.
| | | | - Brian Spencer
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Edward Rockenstein
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Michael Mante
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Paula Desplats
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Wolf Wrasidlo
- Moores Cancer Center, University of California, San Diego, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA.
| | - Anthony Adame
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Emeka Nchekwube
- Immune Modulation, Inc., P.O. Box 998, Bloomington, CA, 92316-0998, USA.
| | - Olusola Oyemade
- Immune Modulation, Inc., P.O. Box 998, Bloomington, CA, 92316-0998, USA.
| | - Joseph Okogun
- Immune Modulation, Inc., P.O. Box 998, Bloomington, CA, 92316-0998, USA.
| | - Michael Chan
- Moores Cancer Center, University of California, San Diego, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA.
| | - Howard Cottam
- Moores Cancer Center, University of California, San Diego, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA. .,Immune Modulation, Inc., P.O. Box 998, Bloomington, CA, 92316-0998, USA.
| | - Eliezer Masliah
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, 92093, USA. .,Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
44
|
Mulak A, Bonaz B. Brain-gut-microbiota axis in Parkinson's disease. World J Gastroenterol 2015; 21:10609-10620. [PMID: 26457021 PMCID: PMC4588083 DOI: 10.3748/wjg.v21.i37.10609] [Citation(s) in RCA: 359] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 05/28/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is characterized by alpha-synucleinopathy that affects all levels of the brain-gut axis including the central, autonomic, and enteric nervous systems. Recently, it has been recognized that the brain-gut axis interactions are significantly modulated by the gut microbiota via immunological, neuroendocrine, and direct neural mechanisms. Dysregulation of the brain-gut-microbiota axis in PD may be associated with gastrointestinal manifestations frequently preceding motor symptoms, as well as with the pathogenesis of PD itself, supporting the hypothesis that the pathological process is spread from the gut to the brain. Excessive stimulation of the innate immune system resulting from gut dysbiosis and/or small intestinal bacterial overgrowth and increased intestinal permeability may induce systemic inflammation, while activation of enteric neurons and enteric glial cells may contribute to the initiation of alpha-synuclein misfolding. Additionally, the adaptive immune system may be disturbed by bacterial proteins cross-reacting with human antigens. A better understanding of the brain-gut-microbiota axis interactions should bring a new insight in the pathophysiology of PD and permit an earlier diagnosis with a focus on peripheral biomarkers within the enteric nervous system. Novel therapeutic options aimed at modifying the gut microbiota composition and enhancing the intestinal epithelial barrier integrity in PD patients could influence the initial step of the following cascade of neurodegeneration in PD.
Collapse
|
45
|
Nazem A, Sankowski R, Bacher M, Al-Abed Y. Rodent models of neuroinflammation for Alzheimer's disease. J Neuroinflammation 2015; 12:74. [PMID: 25890375 PMCID: PMC4404276 DOI: 10.1186/s12974-015-0291-y] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 03/27/2015] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease remains incurable, and the failures of current disease-modifying strategies for Alzheimer's disease could be attributed to a lack of in vivo models that recapitulate the underlying etiology of late-onset Alzheimer's disease. The etiology of late-onset Alzheimer's disease is not based on mutations related to amyloid-β (Aβ) or tau production which are currently the basis of in vivo models of Alzheimer's disease. It has recently been suggested that mechanisms like chronic neuroinflammation may occur prior to amyloid-β and tau pathologies in late-onset Alzheimer's disease. The aim of this study is to analyze the characteristics of rodent models of neuroinflammation in late-onset Alzheimer's disease. Our search criteria were based on characteristics of an idealistic disease model that should recapitulate causes, symptoms, and lesions in a chronological order similar to the actual disease. Therefore, a model based on the inflammation hypothesis of late-onset Alzheimer's disease should include the following features: (i) primary chronic neuroinflammation, (ii) manifestations of memory and cognitive impairment, and (iii) late development of tau and Aβ pathologies. The following models fit the pre-defined criteria: lipopolysaccharide- and PolyI:C-induced models of immune challenge; streptozotocin-, okadaic acid-, and colchicine neurotoxin-induced neuroinflammation models, as well as interleukin-1β, anti-nerve growth factor and p25 transgenic models. Among these models, streptozotocin, PolyI:C-induced, and p25 neuroinflammation models are compatible with the inflammation hypothesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Amir Nazem
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institute for Medical Research, 350 Community drive, Manhasset, NY, 11030, USA.
| | - Roman Sankowski
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institute for Medical Research, 350 Community drive, Manhasset, NY, 11030, USA.
| | - Michael Bacher
- Institute of Immunology, Philipps University Marburg, Hans-Meerwein-Str., 35043, Marburg, Germany.
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, 350 Community drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
46
|
Jiang L, Chen SH, Chu CH, Wang SJ, Oyarzabal E, Wilson B, Sanders V, Xie K, Wang Q, Hong JS. A novel role of microglial NADPH oxidase in mediating extra-synaptic function of norepinephrine in regulating brain immune homeostasis. Glia 2015; 63:1057-72. [PMID: 25740080 DOI: 10.1002/glia.22801] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 01/15/2015] [Indexed: 12/30/2022]
Abstract
Although the peripheral anti-inflammatory effect of norepinephrine (NE) is well documented, the mechanism by which this neurotransmitter functions as an anti-inflammatory/neuroprotective agent in the central nervous system (CNS) is unclear. This article aimed to determine the anti-inflammatory/neuroprotective effects and underlying mechanisms of NE in inflammation-based dopaminergic neurotoxicity models. In mice, NE-depleting toxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) was injected at 6 months of lipopolysaccharide (LPS)-induced neuroinflammation. It was found that NE depletion enhanced LPS-induced dopaminergic neuron loss in the substantia nigra. This piece of in vivo data prompted us to conduct a series of studies in an effort to elucidate the mechanism as to how NE affects dopamine neuron survival by using primary midbrain neuron/glia cultures. Results showed that submicromolar concentrations of NE dose-dependently protected dopaminergic neurons from LPS-induced neurotoxicity by inhibiting microglia activation and subsequent release of pro-inflammatory factors. However, NE-elicited neuroprotection was not totally abolished in cultures from β2-adrenergic receptor (β2-AR)-deficient mice, suggesting that novel pathways other than β2-AR are involved. To this end, It was found that submicromolar NE dose-dependently inhibited NADPH oxidase (NOX2)-generated superoxide, which contributes to the anti-inflammatory and neuroprotective effects of NE. This novel mechanism was indeed adrenergic receptors independent since both (+) and (-) optic isomers of NE displayed the same potency. We further demonstrated that NE inhibited LPS-induced NOX2 activation by blocking the translocation of its cytosolic subunit to plasma membranes. In summary, we revealed a potential physiological role of NE in maintaining brain immune homeostasis and protecting neurons via a novel mechanism.
Collapse
Affiliation(s)
- Lulu Jiang
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, China; Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, 27709
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wang Q, Qian L, Chen SH, Chu CH, Wilson B, Oyarzabal E, Ali S, Robinson B, Rao D, Hong JS. Post-treatment with an ultra-low dose of NADPH oxidase inhibitor diphenyleneiodonium attenuates disease progression in multiple Parkinson's disease models. Brain 2015; 138:1247-62. [PMID: 25716193 DOI: 10.1093/brain/awv034] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/16/2014] [Indexed: 11/13/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate oxidase, a key superoxide-producing enzyme, plays a critical role in microglia-mediated chronic neuroinflammation and subsequent progressive dopaminergic neurodegeneration in Parkinson's disease. Although nicotinamide adenine dinucleotide phosphate oxidase-targeting anti-inflammatory therapy for Parkinson's disease has been proposed, its application in translational research remains limited. The aim of this study was to obtain preclinical evidence supporting this therapeutic strategy by testing the efficacy of an ultra-low dose of the nicotinamide adenine dinucleotide phosphate oxidase inhibitor diphenyleneiodonium in both endotoxin (lipopolysaccharide)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice using post-treatment regimens. Our data revealed that post-treatment with diphenyleneiodonium significantly attenuated progressive dopaminergic degeneration and improved rotarod activity. Remarkably, post-treatment with diphenyleneiodonium 10 months after lipopolysaccharide injection when mice had 30% loss of nigral dopaminergic neurons, showed high efficacy in protecting the remaining neuronal population and restoring motor function. Diphenyleneiodonium-elicited neuroprotection was associated with the inhibition of microglial activation, a reduction in the expression of proinflammatory factors and an attenuation of α-synuclein aggregation. A pathophysiological evaluation of diphenyleneiodonium-treated mice, including assessment of body weight, organs health, and neuronal counts, revealed no overt signs of toxicity. In summary, infusion of ultra-low dose diphenyleneiodonium potently reduced microglia-mediated chronic neuroinflammation by selectively inhibiting nicotinamide adenine dinucleotide phosphate oxidase and halted the progression of neurodegeneration in mouse models of Parkinson's disease. The robust neuroprotective effects and lack of apparent toxic side effects suggest that diphenyleneiodonium at ultra-low dose may be a promising candidate for future clinical trials in Parkinson's disease patients.
Collapse
Affiliation(s)
- Qingshan Wang
- 1 Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Li Qian
- 1 Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Shih-Heng Chen
- 1 Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Chun-Hsien Chu
- 1 Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Belinda Wilson
- 1 Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Esteban Oyarzabal
- 1 Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Syed Ali
- 2 Neurochemistry Laboratory, Division of Neurotoxicology, National Centre for Toxicological Research/USFDA, Jefferson, AR 72079, USA
| | - Bonnie Robinson
- 2 Neurochemistry Laboratory, Division of Neurotoxicology, National Centre for Toxicological Research/USFDA, Jefferson, AR 72079, USA
| | - Deepa Rao
- 3 National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Jau-Shyong Hong
- 1 Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| |
Collapse
|
48
|
Yeh KY, Shou SS, Lin YX, Chen CC, Chiang CY, Yeh CY. Effect of Ginkgo biloba extract on lipopolysaccharide-induced anhedonic depressive-like behavior in male rats. Phytother Res 2014; 29:260-6. [PMID: 25346240 DOI: 10.1002/ptr.5247] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 08/28/2014] [Accepted: 09/28/2014] [Indexed: 11/11/2022]
Abstract
The peripheral administration of lipopolysaccharide (LPS) induces depressive-like behavior. Anhedonia is a core symptom of depression, defined as a loss of the capacity to experience pleasure. The present study used the sucrose preference test to investigate the influence of Ginkgo biloba extract (EGb 761) on LPS-induced anhedonia in male rats. The animals were randomly divided into four groups: (I) vehicle + saline, (II) vehicle + LPS, (III) EGb 761 + saline, and (IV) EGb 761 + LPS. Saline or LPS (100 µg/kg) was administered intraperitoneally 2 h before the sucrose preference test. Sucrose consumption was recorded 2, 4, 6, 13, and 24 h after 100 µg/kg of LPS or saline injection in the dark phase of the light/dark cycle. Dopamine and serotonin levels in the nucleus accumbens were measured. Our results indicated that the vehicle + LPS group exhibited a significant decrease in sucrose intake compared with the vehicle + saline group. The EGb 761 + LPS group showed more sucrose and food consumption than the vehicle + LPS group. Additionally, compared with the EGb 761 + LPS group, the vehicle + LPS group had less dopamine levels in the nucleus accumbens. Treatment with EGb 761 had no effect on water intake. Our results suggest that EGb 761 may be useful for reducing anhedonic depressive-like behavior.
Collapse
Affiliation(s)
- Kuei-Ying Yeh
- Department of Physical Therapy, HungKuang University, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
49
|
Stetler RA, Leak RK, Gan Y, Li P, Zhang F, Hu X, Jing Z, Chen J, Zigmond MJ, Gao Y. Preconditioning provides neuroprotection in models of CNS disease: paradigms and clinical significance. Prog Neurobiol 2014; 114:58-83. [PMID: 24389580 PMCID: PMC3937258 DOI: 10.1016/j.pneurobio.2013.11.005] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/14/2022]
Abstract
Preconditioning is a phenomenon in which brief episodes of a sublethal insult induce robust protection against subsequent lethal injuries. Preconditioning has been observed in multiple organisms and can occur in the brain as well as other tissues. Extensive animal studies suggest that the brain can be preconditioned to resist acute injuries, such as ischemic stroke, neonatal hypoxia/ischemia, surgical brain injury, trauma, and agents that are used in models of neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. Effective preconditioning stimuli are numerous and diverse, ranging from transient ischemia, hypoxia, hyperbaric oxygen, hypothermia and hyperthermia, to exposure to neurotoxins and pharmacological agents. The phenomenon of "cross-tolerance," in which a sublethal stress protects against a different type of injury, suggests that different preconditioning stimuli may confer protection against a wide range of injuries. Research conducted over the past few decades indicates that brain preconditioning is complex, involving multiple effectors such as metabolic inhibition, activation of extra- and intracellular defense mechanisms, a shift in the neuronal excitatory/inhibitory balance, and reduction in inflammatory sequelae. An improved understanding of brain preconditioning should help us identify innovative therapeutic strategies that prevent or at least reduce neuronal damage in susceptible patients. In this review, we focus on the experimental evidence of preconditioning in the brain and systematically survey the models used to develop paradigms for neuroprotection, and then discuss the clinical potential of brain preconditioning.
Collapse
Affiliation(s)
- R Anne Stetler
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Yu Gan
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Peiying Li
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Feng Zhang
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Xiaoming Hu
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Zheng Jing
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Michael J Zigmond
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China.
| |
Collapse
|
50
|
Reinert KRS, Umphlet CD, Quattlebaum A, Boger HA. Short-term effects of an endotoxin on substantia nigra dopamine neurons. Brain Res 2014; 1557:164-70. [PMID: 24513404 DOI: 10.1016/j.brainres.2014.02.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 10/25/2022]
Abstract
Inflammation has been implicated in the pathology of several neurodegenerative diseases, including Parkinson׳s disease (PD). Studies using the endotoxin lipopolysaccharide (LPS), a potent inflammogen, show that systemic insults can trigger prolonged microglial activation and pro-inflammatory cytokine production leading to degeneration of substantia nigra (SN) dopamine (DA) neurons, mimicking idiopathic PD. Because rapid effects of LPS on SN neurons had not been investigated previously, the focus of this study is to assess time-dependent alterations in SN neuroinflammation, DAergic neurons, and neuronal signaling cascades following LPS administration. LPS (5mg/kg, i.p.) or saline (0.9% NaCl) was administered to 8-month-old male mice. At 3h, 5h, and 12h post-injection, the morphology of the SN was assessed using antibodies directed against tyrosine hydroxylase (TH, DAergic marker), Iba-1 (pan-microglial marker), phospho-ERK, and phospho-CREB (signaling). LPS administration significantly reduced TH-immunoreactivity (ir) at all time-points with the greatest reduction observed at 12h post-injection. Reduced TH-ir was accompanied by a significant increase in activated microglia at all time-points following LPS. By 12h post-injection, LPS-treated mice exhibited activated as well as reactive microglia, which can result in neuronal damage. These data demonstrate that the initial reduction in TH-ir observed after an LPS injection was not concomitant with morphological alterations in microglial cells, even though a significant increase in phospho-ERK was observed in glial cells as soon as 3h post-injection. It is possible that the initial alteration in DA phenotype (TH reduction) may perpetuate an inflammatory response that persists and leads to further DAergic damage.
Collapse
Affiliation(s)
- Kaela R S Reinert
- Department of Neurosciences and Center on Aging, Medical University of South Carolina, 173 Ashley Ave, BSB Suite 403, MSC 510, Charleston, SC 29425, USA
| | - Claudia D Umphlet
- Department of Neurosciences and Center on Aging, Medical University of South Carolina, 173 Ashley Ave, BSB Suite 403, MSC 510, Charleston, SC 29425, USA
| | - Ariana Quattlebaum
- Department of Neurosciences and Center on Aging, Medical University of South Carolina, 173 Ashley Ave, BSB Suite 403, MSC 510, Charleston, SC 29425, USA
| | - Heather A Boger
- Department of Neurosciences and Center on Aging, Medical University of South Carolina, 173 Ashley Ave, BSB Suite 403, MSC 510, Charleston, SC 29425, USA.
| |
Collapse
|