1
|
Trainor AR, MacDonald DS, Penney J. Microglia: roles and genetic risk in Parkinson's disease. Front Neurosci 2024; 18:1506358. [PMID: 39554849 PMCID: PMC11564156 DOI: 10.3389/fnins.2024.1506358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
The prevalence of neurodegenerative disorders such as Parkinson's disease are increasing as world populations age. Despite this growing public health concern, the precise molecular and cellular mechanisms that culminate in neurodegeneration remain unclear. Effective treatment options for Parkinson's disease and other neurodegenerative disorders remain very limited, due in part to this uncertain disease etiology. One commonality across neurodegenerative diseases is sustained neuroinflammation, mediated in large part by microglia, the innate immune cells of the brain. Initially thought to simply react to neuron-derived pathology, genetic and functional studies in recent years suggest that microglia play a more active role in the neurodegenerative process than previously appreciated. Here, we review evidence for the roles of microglia in Parkinson's disease pathogenesis and progression, with a particular focus on microglial functions that are perturbed by disease associated genes and mutations.
Collapse
Affiliation(s)
| | | | - Jay Penney
- Department of Biomedical Sciences, AVC, University of Prince Edward Island, Charlottetown, PE, Canada
| |
Collapse
|
2
|
Pan H, Huang M, Zhu C, Lin S, He L, Shen R, Chen Y, Fang F, Qiu Y, Qin M, Bao P, Tan Y, Xu J, Ding J, Chen S. A novel compound alleviates oxidative stress via PKA/CREB1-mediated DJ-1 upregulation. J Neurochem 2024; 168:3034-3049. [PMID: 38994800 DOI: 10.1111/jnc.16161] [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: 06/10/2023] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 07/13/2024]
Abstract
Oxidative stress is one of the major culprits causing dopaminergic neuron loss in Parkinson's disease (PD). DJ-1 is a protein with multiple actions against oxidative stress, apoptosis, neuroinflammation, etc. DJ-1 expression is decreased in sporadic PD, therefore increasing DJ-1 expression might be beneficial in PD treatment. However, drugs known to upregulate DJ-1 are still lacking. In this study, we identified a novel DJ-1-elevating compound called ChemJ through luciferase assay-based high-throughput compound screening in SH-SY5Y cells and confirmed that ChemJ upregulated DJ-1 in SH-SY5Y cell line and primary cortical neurons. DJ-1 upregulation by ChemJ alleviated MPP+-induced oxidative stress. In exploring the underlying mechanisms, we found that the transcription factor CREB1 bound to DJ-1 promoter and positively regulated its expression under both unstressed and 1-methyl-4-phenylpyridinium-induced oxidative stress conditions and that ChemJ promoted DJ-1 expression via activating PKA/CREB1 pathway in SH-SY5Y cells. Our results demonstrated that ChemJ alleviated the MPP+-induced oxidative stress through a PKA/CREB1-mediated regulation of DJ-1 expression, thus offering a novel and promising avenue for PD treatment.
Collapse
Affiliation(s)
- Hong Pan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, China
| | - Maoxin Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenxiang Zhu
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, China
| | - Suzhen Lin
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu He
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruinan Shen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimeng Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Fang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinghui Qiu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meiling Qin
- Institute of Neuroscience and State key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Puhua Bao
- Institute of Neuroscience and State key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Yuyan Tan
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Xu
- Institute of Neuroscience and State key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Jianqing Ding
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, China
| |
Collapse
|
3
|
Lind-Holm Mogensen F, Sousa C, Ameli C, Badanjak K, Pereira SL, Muller A, Kaoma T, Coowar D, Scafidi A, Poovathingal SK, Tziortziou M, Antony PMA, Nicot N, Ginolhac A, Vogt Weisenhorn DM, Wurst W, Poli A, Nazarov PV, Skupin A, Grünewald A, Michelucci A. PARK7/DJ-1 deficiency impairs microglial activation in response to LPS-induced inflammation. J Neuroinflammation 2024; 21:174. [PMID: 39014482 PMCID: PMC11253405 DOI: 10.1186/s12974-024-03164-x] [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: 02/05/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Specific microglia responses are thought to contribute to the development and progression of neurodegenerative diseases, including Parkinson's disease (PD). However, the phenotypic acquisition of microglial cells and their role during the underlying neuroinflammatory processes remain largely elusive. Here, according to the multiple-hit hypothesis, which stipulates that PD etiology is determined by a combination of genetics and various environmental risk factors, we investigate microglial transcriptional programs and morphological adaptations under PARK7/DJ-1 deficiency, a genetic cause of PD, during lipopolysaccharide (LPS)-induced inflammation. METHODS Using a combination of single-cell RNA-sequencing, bulk RNA-sequencing, multicolor flow cytometry and immunofluorescence analyses, we comprehensively compared microglial cell phenotypic characteristics in PARK7/DJ-1 knock-out (KO) with wildtype littermate mice following 6- or 24-h intraperitoneal injection with LPS. For translational perspectives, we conducted corresponding analyses in human PARK7/DJ-1 mutant induced pluripotent stem cell (iPSC)-derived microglia and murine bone marrow-derived macrophages (BMDMs). RESULTS By excluding the contribution of other immune brain resident and peripheral cells, we show that microglia acutely isolated from PARK7/DJ-1 KO mice display a distinct phenotype, specially related to type II interferon and DNA damage response signaling, when compared with wildtype microglia, in response to LPS. We also detected discrete signatures in human PARK7/DJ-1 mutant iPSC-derived microglia and BMDMs from PARK7/DJ-1 KO mice. These specific transcriptional signatures were reflected at the morphological level, with microglia in LPS-treated PARK7/DJ-1 KO mice showing a less amoeboid cell shape compared to wildtype mice, both at 6 and 24 h after acute inflammation, as also observed in BMDMs. CONCLUSIONS Taken together, our results show that, under inflammatory conditions, PARK7/DJ-1 deficiency skews microglia towards a distinct phenotype characterized by downregulation of genes involved in type II interferon signaling and a less prominent amoeboid morphology compared to wildtype microglia. These findings suggest that the underlying oxidative stress associated with the lack of PARK7/DJ-1 affects microglia neuroinflammatory responses, which may play a causative role in PD onset and progression.
Collapse
Affiliation(s)
- Frida Lind-Holm Mogensen
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Carole Sousa
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- International Iberian Nanotechnology Laboratory, 4715-330, Braga, Portugal
| | - Corrado Ameli
- Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Katja Badanjak
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Sandro L Pereira
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Arnaud Muller
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- LuxGen Genome Center, Luxembourg Institute of Health and Laboratoire National de Santé, L-3555, Dudelange, Luxembourg
| | - Tony Kaoma
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Djalil Coowar
- Rodent Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Andrea Scafidi
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Suresh K Poovathingal
- Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Single Cell Analytics and Microfluidics Core, Vlaams Instituut Voor Biotechnologie-KU Leuven, 3000, Louvain, Belgium
| | - Maria Tziortziou
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Paul M A Antony
- Bioimaging Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
| | - Nathalie Nicot
- LuxGen Genome Center, Luxembourg Institute of Health and Laboratoire National de Santé, L-3555, Dudelange, Luxembourg
| | - Aurélien Ginolhac
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Daniela M Vogt Weisenhorn
- Institute of Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Technische Universität München-Weihenstephan, 85354, Freising, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Technische Universität München-Weihenstephan, 85354, Freising, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
- Deutsche Zentrum für Psychische Gesundheit (DZPG), 80336, Munich, Germany
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Petr V Nazarov
- Bioinformatics Platform, Department of Medical Informatics, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
- Multiomics Data Science Group, Department of Cancer Research, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg
| | - Alexander Skupin
- Integrative Cell Signalling Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Department of Neuroscience, University of California San Diego, La Jolla, CA, 92093, USA
- Integrative Biophysics, Department of Physics and Material Science, University of Luxembourg, L-1511, Luxembourg, Luxembourg
| | - Anne Grünewald
- Molecular and Functional Neurobiology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362, Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, 23538, Lübeck, Germany
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A, rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg.
| |
Collapse
|
4
|
Bharadwaj S, Groza Y, Mierzwicka JM, Malý P. Current understanding on TREM-2 molecular biology and physiopathological functions. Int Immunopharmacol 2024; 134:112042. [PMID: 38703564 DOI: 10.1016/j.intimp.2024.112042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 05/06/2024]
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM-2), a glycosylated receptor belonging to the immunoglobin superfamily and especially expressed in the myeloid cell lineage, is frequently explained as a reminiscent receptor for both adaptive and innate immunity regulation. TREM-2 is also acknowledged to influence NK cell differentiation via the PI3K and PLCγ signaling pathways, as well as the partial activation or direct inhibition of T cells. Additionally, TREM-2 overexpression is substantially linked to cell-specific functions, such as enhanced phagocytosis, reduced toll-like receptor (TLR)-mediated inflammatory cytokine production, increased transcription of anti-inflammatory cytokines, and reshaped T cell function. Whereas TREM-2-deficient cells exhibit diminished phagocytic function and enhanced proinflammatory cytokines production, proceeding to inflammatory injuries and an immunosuppressive environment for disease progression. Despite the growing literature supporting TREM-2+ cells in various diseases, such as neurodegenerative disorders and cancer, substantial facets of TREM-2-mediated signaling remain inadequately understood relevant to pathophysiology conditions. In this direction, herein, we have summarized the current knowledge on TREM-2 biology and cell-specific TREM-2 expression, particularly in the modulation of pivotal TREM-2-dependent functions under physiopathological conditions. Furthermore, molecular regulation and generic biological relevance of TREM-2 are also discussed, which might provide an alternative approach for preventing or reducing TREM-2-associated deformities. At last, we discussed the TREM-2 function in supporting an immunosuppressive cancer environment and as a potential drug target for cancer immunotherapy. Hence, summarized knowledge of TREM-2 might provide a window to overcome challenges in clinically effective therapies for TREM-2-induced diseases in humans.
Collapse
Affiliation(s)
- Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic.
| | - Yaroslava Groza
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Joanna M Mierzwicka
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic.
| |
Collapse
|
5
|
Albertini C, Petralla S, Massenzio F, Monti B, Rizzardi N, Bergamini C, Uliassi E, Borges F, Chavarria D, Fricker G, Goettert M, Kronenberger T, Gehringer M, Laufer S, Bolognesi ML. Targeting Lewy body dementia with neflamapimod-rasagiline hybrids. Arch Pharm (Weinheim) 2024; 357:e2300525. [PMID: 38412454 DOI: 10.1002/ardp.202300525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024]
Abstract
Lewy body dementia (LBD) represents the second most common neurodegenerative dementia but is a quite underexplored therapeutic area. Nepflamapimod (1) is a brain-penetrant selective inhibitor of the alpha isoform of the mitogen-activated serine/threonine protein kinase (MAPK) p38α, recently repurposed for LBD due to its remarkable antineuroinflammatory properties. Neuroprotective propargylamines are another class of molecules with a therapeutical potential against LBD. Herein, we sought to combine the antineuroinflammatory core of 1 and the neuroprotective propargylamine moiety into a single molecule. Particularly, we inserted a propargylamine moiety in position 4 of the 2,6-dichlorophenyl ring of 1, generating neflamapimod-propargylamine hybrids 3 and 4. These hybrids were evaluated using several cell models, aiming to recapitulate the complexity of LBD pathology through different molecular mechanisms. The N-methyl-N-propargyl derivative 4 showed a nanomolar p38α-MAPK inhibitory activity (IC50 = 98.7 nM), which is only 2.6-fold lower compared to that of the parent compound 1, while displaying no hepato- and neurotoxicity up to 25 μM concentration. It also retained a similar immunomodulatory profile against the N9 microglial cell line. Gratifyingly, at 5 μM concentration, 4 demonstrated a neuroprotective effect against dexamethasone-induced reactive oxygen species production in neuronal cells that was higher than that of 1.
Collapse
Affiliation(s)
- Claudia Albertini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Sabrina Petralla
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Francesca Massenzio
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Nicola Rizzardi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Fernanda Borges
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Marcia Goettert
- Department of Pharmaceutical and Medicinal Chemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Thales Kronenberger
- Department of Pharmaceutical and Medicinal Chemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Matthias Gehringer
- Department of Pharmaceutical and Medicinal Chemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Maria L Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
6
|
Eser P, Kocabicak E, Bekar A, Temel Y. The interplay between neuroinflammatory pathways and Parkinson's disease. Exp Neurol 2024; 372:114644. [PMID: 38061555 DOI: 10.1016/j.expneurol.2023.114644] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Parkinson's disease, a progressive neurodegenerative disorder predominantly affecting elderly, is marked by the gradual degeneration of the nigrostriatal dopaminergic pathway, culminating in neuronal loss within the substantia nigra pars compacta (SNpc) and dopamine depletion. At the molecular level, neuronal loss in the SNpc has been attributed to factors including neuroinflammation, impaired protein homeostasis, as well as mitochondrial dysfunction and the resulting oxidative stress. This review focuses on the interplay between neuroinflammatory pathways and Parkinson's disease, drawing insights from current literature.
Collapse
Affiliation(s)
- Pinar Eser
- Bursa Uludag University School of Medicine, Department of Neurosurgery, Bursa, Turkey.
| | - Ersoy Kocabicak
- Ondokuz Mayis University, Health Practise and Research Hospital, Neuromodulation Center, Samsun, Turkey
| | - Ahmet Bekar
- Bursa Uludag University School of Medicine, Department of Neurosurgery, Bursa, Turkey
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
7
|
Xue J, Tao K, Wang W, Wang X. What Can Inflammation Tell Us about Therapeutic Strategies for Parkinson's Disease? Int J Mol Sci 2024; 25:1641. [PMID: 38338925 PMCID: PMC10855787 DOI: 10.3390/ijms25031641] [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: 01/01/2024] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder with a complicated etiology and pathogenesis. α-Synuclein aggregation, dopaminergic (DA) neuron loss, mitochondrial injury, oxidative stress, and inflammation are involved in the process of PD. Neuroinflammation has been recognized as a key element in the initiation and progression of PD. In this review, we summarize the inflammatory response and pathogenic mechanisms of PD. Additionally, we describe the potential anti-inflammatory therapies, including nod-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome inhibition, nuclear factor κB (NF-κB) inhibition, microglia inhibition, astrocyte inhibition, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibition, the peroxisome proliferator-activated receptor γ (PPARγ) agonist, targeting the mitogen-activated protein kinase (MAPK) pathway, targeting the adenosine monophosphate-activated protein kinase (AMPK)-dependent pathway, targeting α-synuclein, targeting miRNA, acupuncture, and exercise. The review focuses on inflammation and will help in designing new prevention strategies for PD.
Collapse
Affiliation(s)
- Jinsong Xue
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| | | | | | - Xiaofei Wang
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| |
Collapse
|
8
|
Zhang X, Chen X, Zhang L, Sun Y, Liang Y, Li H, Zhang Y. Role of trigger receptor 2 expressed on myeloid cells in neuroinflammation-neglected multidimensional regulation of microglia. Neurochem Int 2023; 171:105639. [PMID: 37926352 DOI: 10.1016/j.neuint.2023.105639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/01/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023]
Abstract
Neuroinflammation is an inflammatory cascade involved in various neurological disorders, including Alzheimer's disease, multiple sclerosis, and other relevant diseases. The triggering receptor expressed on myeloid cells 2 (TREM2) is a transmembrane immune receptor that is primarily expressed by microglia in the central nervous system (CNS). While TREM2 is initially believed to be an anti-inflammatory factor in the CNS, increasing evidence suggests that TREM2 plays a more complex role in balancing neuroinflammation. However, the exact mechanism remains unclear. Notably, TREM2 directly regulates microglia inflammation through various signaling pathways. Additionally, studies have suggested that TREM2 mediates microglial phagocytosis, autophagy, metabolism, and microglia phenotypes, which may be involved in the modulation of neuroinflammation. In this review, we aim to discuss the critical role of TREM2 in several microglia functions and the underlying molecular mechanism the modulatory which further mediate neuroinflammation, and elaborate. Finally, we discuss the potential of TREM2 as a therapeutic target in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China; Beijing Institute of Hepatology, Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xue Chen
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China; Beijing Institute of Hepatology, Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ling Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yuqing Sun
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ying Liang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Huan Li
- Department of Cardiology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China; Beijing Institute of Hepatology, Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
9
|
Liu LL, Han Y, Zhang ZJ, Wang YQ, Hu YW, Kaznacheyeva E, Ding JQ, Guo DK, Wang GH, Li B, Ren HG. Loss of DJ-1 function contributes to Parkinson's disease pathogenesis in mice via RACK1-mediated PKC activation and MAO-B upregulation. Acta Pharmacol Sin 2023; 44:1948-1961. [PMID: 37225849 PMCID: PMC10545772 DOI: 10.1038/s41401-023-01104-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/01/2023] [Indexed: 05/26/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative motor disorder characterized by a dramatic reduction in pars compacta of substantia nigra dopaminergic neurons and striatal dopamine (DA) levels. Mutations or deletions in the PARK7/DJ-1 gene are associated with an early-onset familial form of PD. DJ-1 protein prevents neurodegeneration via its regulation of oxidative stress and mitochondrial function as well as its roles in transcription and signal transduction. In this study, we investigated how loss of DJ-1 function affected DA degradation, ROS generation and mitochondrial dysfunction in neuronal cells. We showed that loss of DJ-1 significantly increased the expression of monoamine oxidase (MAO)-B but not MAO-A in both neuronal cells and primary astrocytes. In DJ-1-knockout (KO) mice, MAO-B protein levels in the substantia nigra (SN) and striatal regions were significantly increased. We demonstrated that the induction of MAO-B expression by DJ-1 deficiency depended on early growth response 1 (EGR1) in N2a cells. By coimmunoprecipitation omics analysis, we found that DJ-1 interacted with receptor of activated protein C kinase 1 (RACK1), a scaffolding protein, and thus inhibited the activity of the PKC/JNK/AP-1/EGR1 cascade. The PKC inhibitor sotrastaurin or the JNK inhibitor SP600125 completely inhibited DJ-1 deficiency-induced EGR1 and MAO-B expression in N2a cells. Moreover, the MAO-B inhibitor rasagiline inhibited mitochondrial ROS generation and rescued neuronal cell death caused by DJ-1 deficiency, especially in response to MPTP stimulation in vitro and in vivo. These results suggest that DJ-1 exerts neuroprotective effects by inhibiting the expression of MAO-B distributed at the mitochondrial outer membrane, which mediates DA degradation, ROS generation and mitochondrial dysfunction. This study reveals a mechanistic link between DJ-1 and MAO-B expression and contributes to understanding the crosslinks among pathogenic factors, mitochondrial dysfunction and oxidative stress in PD pathogenesis.
Collapse
Affiliation(s)
- Le-le Liu
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yu Han
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zi-Jia Zhang
- Qingdao Municipal Hospital of Shandong Province, Qingdao, 266011, China
| | - Yi-Qi Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yu-Wei Hu
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Elena Kaznacheyeva
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, 194064, Russia
| | - Jian-Qing Ding
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Dong-Kai Guo
- Laboratory of Clinical Pharmacy, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
| | - Guang-Hui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Bin Li
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, 215200, China.
| | - Hai-Gang Ren
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
10
|
Jurcau A, Andronie-Cioara FL, Nistor-Cseppento DC, Pascalau N, Rus M, Vasca E, Jurcau MC. The Involvement of Neuroinflammation in the Onset and Progression of Parkinson's Disease. Int J Mol Sci 2023; 24:14582. [PMID: 37834030 PMCID: PMC10573049 DOI: 10.3390/ijms241914582] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/15/2023] Open
Abstract
Parkinson's disease is a neurodegenerative disease exhibiting the fastest growth in incidence in recent years. As with most neurodegenerative diseases, the pathophysiology is incompletely elucidated, but compelling evidence implicates inflammation, both in the central nervous system and in the periphery, in the initiation and progression of the disease, although it is not yet clear what triggers this inflammatory response and where it begins. Gut dysbiosis seems to be a likely candidate for the initiation of the systemic inflammation. The therapies in current use provide only symptomatic relief, but do not interfere with the disease progression. Nonetheless, animal models have shown promising results with therapies that target various vicious neuroinflammatory cascades. Translating these therapeutic strategies into clinical trials is still in its infancy, and a series of issues, such as the exact timing, identifying biomarkers able to identify Parkinson's disease in early and pre-symptomatic stages, or the proper indications of genetic testing in the population at large, will need to be settled in future guidelines.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (A.J.); (D.C.N.-C.)
| | - Felicia Liana Andronie-Cioara
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (A.J.); (D.C.N.-C.)
| | - Delia Carmen Nistor-Cseppento
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (A.J.); (D.C.N.-C.)
| | - Nicoleta Pascalau
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (A.J.); (D.C.N.-C.)
| | - Marius Rus
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Elisabeta Vasca
- Department of Oral Rehabilitation, Faculty of Medicine “Vasile Goldis” Arad, 310025 Arad, Romania
| | | |
Collapse
|
11
|
Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther 2023; 8:359. [PMID: 37735487 PMCID: PMC10514343 DOI: 10.1038/s41392-023-01588-0] [Citation(s) in RCA: 294] [Impact Index Per Article: 147.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 09/23/2023] Open
Abstract
Microglia activation is observed in various neurodegenerative diseases. Recent advances in single-cell technologies have revealed that these reactive microglia were with high spatial and temporal heterogeneity. Some identified microglia in specific states correlate with pathological hallmarks and are associated with specific functions. Microglia both exert protective function by phagocytosing and clearing pathological protein aggregates and play detrimental roles due to excessive uptake of protein aggregates, which would lead to microglial phagocytic ability impairment, neuroinflammation, and eventually neurodegeneration. In addition, peripheral immune cells infiltration shapes microglia into a pro-inflammatory phenotype and accelerates disease progression. Microglia also act as a mobile vehicle to propagate protein aggregates. Extracellular vesicles released from microglia and autophagy impairment in microglia all contribute to pathological progression and neurodegeneration. Thus, enhancing microglial phagocytosis, reducing microglial-mediated neuroinflammation, inhibiting microglial exosome synthesis and secretion, and promoting microglial conversion into a protective phenotype are considered to be promising strategies for the therapy of neurodegenerative diseases. Here we comprehensively review the biology of microglia and the roles of microglia in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, multiple system atrophy, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration, dementia with Lewy bodies and Huntington's disease. We also summarize the possible microglia-targeted interventions and treatments against neurodegenerative diseases with preclinical and clinical evidence in cell experiments, animal studies, and clinical trials.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jingwen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
12
|
Lind-Holm Mogensen F, Scafidi A, Poli A, Michelucci A. PARK7/DJ-1 in microglia: implications in Parkinson's disease and relevance as a therapeutic target. J Neuroinflammation 2023; 20:95. [PMID: 37072827 PMCID: PMC10111685 DOI: 10.1186/s12974-023-02776-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/11/2023] [Indexed: 04/20/2023] Open
Abstract
Microglia are the immune effector cells of the brain playing critical roles in immune surveillance and neuroprotection in healthy conditions, while they can sustain neuroinflammatory and neurotoxic processes in neurodegenerative diseases, including Parkinson's disease (PD). Although the precise triggers of PD remain obscure, causative genetic mutations, which aid in the identification of molecular pathways underlying the pathogenesis of idiopathic forms, represent 10% of the patients. Among the inherited forms, loss of function of PARK7, which encodes the protein DJ-1, results in autosomal recessive early-onset PD. Yet, although protection against oxidative stress is the most prominent task ascribed to DJ-1, the underlying mechanisms linking DJ-1 deficiency to the onset of PD are a current matter of investigation. This review provides an overview of the role of DJ-1 in neuroinflammation, with a special focus on its functions in microglia genetic programs and immunological traits. Furthermore, it discusses the relevance of targeting dysregulated pathways in microglia under DJ-1 deficiency and their importance as therapeutic targets in PD. Lastly, it addresses the prospect to consider DJ-1, detected in its oxidized form in idiopathic PD, as a biomarker and to take into account DJ-1-enhancing compounds as therapeutics dampening oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Frida Lind-Holm Mogensen
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Doctoral School of Science and Technology, University of Luxembourg, 7 Avenue Des Haut Forneuaux, L-4362, Esch-Sur-Alzette, Luxembourg
| | - Andrea Scafidi
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Doctoral School of Science and Technology, University of Luxembourg, 7 Avenue Des Haut Forneuaux, L-4362, Esch-Sur-Alzette, Luxembourg
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg.
| |
Collapse
|
13
|
Isik S, Yeman Kiyak B, Akbayir R, Seyhali R, Arpaci T. Microglia Mediated Neuroinflammation in Parkinson’s Disease. Cells 2023; 12:cells12071012. [PMID: 37048085 PMCID: PMC10093562 DOI: 10.3390/cells12071012] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Parkinson’s Disease (PD) is the second most common neurodegenerative disorder seen, especially in the elderly. Tremor, shaking, movement problems, and difficulty with balance and coordination are among the hallmarks, and dopaminergic neuronal loss in substantia nigra pars compacta of the brain and aggregation of intracellular protein α-synuclein are the pathological characterizations. Neuroinflammation has emerged as an involving mechanism at the initiation and development of PD. It is a complex network of interactions comprising immune and non-immune cells in addition to mediators of the immune response. Microglia, the resident macrophages in the CNS, take on the leading role in regulating neuroinflammation and maintaining homeostasis. Under normal physiological conditions, they exist as “homeostatic” but upon pathological stimuli, they switch to the “reactive state”. Pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes are used to classify microglial activity with each phenotype having its own markers and released mediators. When M1 microglia are persistent, they will contribute to various inflammatory diseases, including neurodegenerative diseases, such as PD. In this review, we focus on the role of microglia mediated neuroinflammation in PD and also signaling pathways, receptors, and mediators involved in the process, presenting the studies that associate microglia-mediated inflammation with PD. A better understanding of this complex network and interactions is important in seeking new therapies for PD and possibly other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sevim Isik
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Uskudar, Istanbul 34662, Turkey
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Correspondence: ; Tel.: +90-216-400-2222 (ext. 2462)
| | - Bercem Yeman Kiyak
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Medicine, Institute of Hamidiye Health Sciences, University of Health Sciences, Uskudar, Istanbul 34668, Turkey
| | - Rumeysa Akbayir
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Rama Seyhali
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Tahire Arpaci
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| |
Collapse
|
14
|
Lv QK, Tao KX, Wang XB, Yao XY, Pang MZ, Liu JY, Wang F, Liu CF. Role of α-synuclein in microglia: autophagy and phagocytosis balance neuroinflammation in Parkinson's disease. Inflamm Res 2023; 72:443-462. [PMID: 36598534 DOI: 10.1007/s00011-022-01676-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/27/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disease, and is characterized by accumulation of α-synuclein (α-syn). Neuroinflammation driven by microglia is an important pathological manifestation of PD. α-Syn is a crucial marker of PD, and its accumulation leads to microglia M1-like phenotype polarization, activation of NLRP3 inflammasomes, and impaired autophagy and phagocytosis in microglia. Autophagy of microglia is related to degradation of α-syn and NLRP3 inflammasome blockage to relieve neuroinflammation. Microglial autophagy and phagocytosis of released α-syn or fragments from apoptotic neurons maintain homeostasis in the brain. A variety of PD-related genes such as LRRK2, GBA and DJ-1 also contribute to this stability process. OBJECTIVES Further studies are needed to determine how α-syn works in microglia. METHODS A keyword-based search was performed using the PubMed database for published articles. CONCLUSION In this review, we discuss the interaction between microglia and α-syn in PD pathogenesis and the possible mechanism of microglial autophagy and phagocytosis in α-syn clearance and inhibition of neuroinflammation. This may provide a novel insight into treatment of PD.
Collapse
Affiliation(s)
- Qian-Kun Lv
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Kang-Xin Tao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiao-Bo Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiao-Yu Yao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Meng-Zhu Pang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Jun-Yi Liu
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
15
|
Recent Advances in the Treatment of Genetic Forms of Parkinson's Disease: Hype or Hope? Cells 2023; 12:cells12050764. [PMID: 36899899 PMCID: PMC10001341 DOI: 10.3390/cells12050764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Parkinson's disease (PD) is a multifarious neurodegenerative disease. Its pathology is characterized by a prominent early death of dopaminergic neurons in the pars compacta of the substantia nigra and the presence of Lewy bodies with aggregated α-synuclein. Although the α-synuclein pathological aggregation and propagation, induced by several factors, is considered one of the most relevant hypotheses, PD pathogenesis is still a matter of debate. Indeed, environmental factors and genetic predisposition play an important role in PD. Mutations associated with a high risk for PD, usually called monogenic PD, underlie 5% to 10% of all PD cases. However, this percentage tends to increase over time because of the continuous identification of new genes associated with PD. The identification of genetic variants that can cause or increase the risk of PD has also given researchers the possibility to explore new personalized therapies. In this narrative review, we discuss the recent advances in the treatment of genetic forms of PD, focusing on different pathophysiologic aspects and ongoing clinical trials.
Collapse
|
16
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
17
|
Mechanisms of Autoimmune Cell in DA Neuron Apoptosis of Parkinson's Disease: Recent Advancement. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7965433. [PMID: 36567855 PMCID: PMC9771667 DOI: 10.1155/2022/7965433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder that manifests as motor and nonmotor symptoms due to the selective loss of midbrain DArgic (DA) neurons. More and more studies have shown that pathological reactions initiated by autoimmune cells play an essential role in the progression of PD. Autoimmune cells exist in the brain parenchyma, cerebrospinal fluid, and meninges; they are considered inducers of neuroinflammation and regulate the immune in the human brain in PD. For example, T cells can recognize α-synuclein presented by antigen-presenting cells to promote neuroinflammation. In addition, B cells will accelerate the apoptosis of DA neurons in the case of PD-related gene mutations. Activation of microglia and damage of DA neurons even form the self-degeneration cycle to deteriorate PD. Numerous autoimmune cells have been considered regulators of apoptosis, α-synuclein misfolding and aggregation, mitochondrial dysfunction, autophagy, and neuroinflammation of DA neurons in PD. The evidence is mounting that autoimmune cells promote DA neuron apoptosis. In this review, we discuss the current knowledge regarding the regulation and function of B cell, T cell, and microglia as well as NK cell in PD pathogenesis, focusing on DA neuron apoptosis to understand the disease better and propose potential target identification for the treatment in the early stages of PD. However, there are still some limitations in our work, for example, the specific mechanism of PD progression caused by autoimmune cells in mitochondrial dysfunction, ferroptosis, and autophagy has not been clarified in detail, which needs to be summarized in further work.
Collapse
|
18
|
Naoi M, Maruyama W, Shamoto-Nagai M. Neuroprotective Function of Rasagiline and Selegiline, Inhibitors of Type B Monoamine Oxidase, and Role of Monoamine Oxidases in Synucleinopathies. Int J Mol Sci 2022; 23:ijms231911059. [PMID: 36232361 PMCID: PMC9570229 DOI: 10.3390/ijms231911059] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/27/2022] Open
Abstract
Synucleinopathies are a group of neurodegenerative disorders caused by the accumulation of toxic species of α-synuclein. The common clinical features are chronic progressive decline of motor, cognitive, behavioral, and autonomic functions. They include Parkinson’s disease, dementia with Lewy body, and multiple system atrophy. Their etiology has not been clarified and multiple pathogenic factors include oxidative stress, mitochondrial dysfunction, impaired protein degradation systems, and neuroinflammation. Current available therapy cannot prevent progressive neurodegeneration and “disease-modifying or neuroprotective” therapy has been proposed. This paper presents the molecular mechanisms of neuroprotection by the inhibitors of type B monoamine oxidase, rasagiline and selegiline. They prevent mitochondrial apoptosis, induce anti-apoptotic Bcl-2 protein family, and pro-survival brain- and glial cell line-derived neurotrophic factors. They also prevent toxic oligomerization and aggregation of α-synuclein. Monoamine oxidase is involved in neurodegeneration and neuroprotection, independently of the catalytic activity. Type A monoamine oxidases mediates rasagiline-activated signaling pathways to induce neuroprotective genes in neuronal cells. Multi-targeting propargylamine derivatives have been developed for therapy in various neurodegenerative diseases. Preclinical studies have presented neuroprotection of rasagiline and selegiline, but beneficial effects have been scarcely presented. Strategy to improve clinical trials is discussed to achieve disease-modification in synucleinopathies.
Collapse
Affiliation(s)
- Makoto Naoi
- Correspondence: ; Tel.: +81-05-6173-1111 (ext. 3494); Fax: +81-561-731-142
| | | | | |
Collapse
|
19
|
Cunha DMG, Becegato M, Meurer YSR, Lima AC, Gonçalves N, Bioni VS, Engi SA, Bianchi PC, Cruz FC, Santos JR, Silva RH. Neuroinflammation in early, late and recovery stages in a progressive parkinsonism model in rats. Front Neurosci 2022; 16:923957. [PMID: 36090265 PMCID: PMC9459164 DOI: 10.3389/fnins.2022.923957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by motor and non-motor signs, which are accompanied by progressive degeneration of dopaminergic neurons in the substantia nigra. Although the exact causes are unknown, evidence links this neuronal loss with neuroinflammation and oxidative stress. Repeated treatment with a low dose of reserpine—inhibitor of VMAT2—has been proposed as a progressive pharmacological model of PD. The aim of this study was to investigate whether this model replicates the neuroinflammation characteristic of this disease. Six-month-old Wistar rats received repeated subcutaneous injections of reserpine (0.1 mg/kg) or vehicle on alternate days. Animals were euthanized after 5, 10, or 15 injections, or 20 days after the 15th injection. Catalepsy tests (motor assessment) were conducted across treatment. Brains were collected at the end of each treatment period for immunohistochemical and RT-PCR analyzes. Reserpine induced a significant progressive increase in catalepsy duration. We also found decreased immunostaining for tyrosine hydroxylase (TH) in the substantia nigra pars compacta (SNpc) and increased GFAP + cells in the SNpc and dorsal striatum after 10 and 15 reserpine injections. Phenotyping microglial M1 and M2 markers showed increased number of CD11b + cells and percentage of CD11b + /iNOS + cells in reserpine-treated animals after 15 injections, which is compatible with tissue damage and production of cytotoxic factors. In addition, increased CD11b + /ArgI + cells were found 20 days after the last reserpine injection, together with an increment in IL-10 gene expression in the dorsal striatum, which is indicative of tissue repair or regeneration. Reserpine also induced increases in striatal interleukin TNF-alpha mRNA levels in early stages. In view of these results, we conclude that reserpine-induced progressive parkinsonism model leads to neuroinflammation in regions involved in the pathophysiology of PD, which is reversed 20 days after the last injection. These findings reveal that withdrawal period, together with the shift of microglial phenotypes from the pro-inflammatory to the anti-inflammatory stage, may be important for the study of the mechanisms involved in reversing this condition, with potential clinical applicability.
Collapse
Affiliation(s)
- Debora M. G. Cunha
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marcela Becegato
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ywlliane S. R. Meurer
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alvaro C. Lima
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Narriman Gonçalves
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Vinícius S. Bioni
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sheila A. Engi
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Paula C. Bianchi
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fabio C. Cruz
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jose R. Santos
- Behavioral and Evolutionary Neurobiology Laboratory, Department of Bioscience, Universidade Federal do Sergipe, Itabaiana, Brazil
| | - Regina H. Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
- *Correspondence: Regina H. Silva,
| |
Collapse
|
20
|
Neurotransmitters: Potential Targets in Glioblastoma. Cancers (Basel) 2022; 14:cancers14163970. [PMID: 36010960 PMCID: PMC9406056 DOI: 10.3390/cancers14163970] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/01/2022] [Accepted: 08/12/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Aiming to discover potential treatments for GBM, this review connects emerging research on the roles of neurotransmitters in the normal neural and the GBM microenvironments and sheds light on the prospects of their application in the neuropharmacology of GBM. Conventional therapy is blamed for its poor effect, especially in inhibiting tumor recurrence and invasion. Facing this dilemma, we focus on neurotransmitters that modulate GBM initiation, progression and invasion, hoping to provide novel therapy targeting GBM. By analyzing research concerning GBM therapy systematically and scientifically, we discover increasing insights into the regulatory effects of neurotransmitters, some of which have already shown great potential in research in vivo or in vitro. After that, we further summarize the potential drugs in correlation with previously published research. In summary, it is worth expecting that targeting neurotransmitters could be a promising novel pharmacological approach for GBM treatment. Abstract For decades, glioblastoma multiforme (GBM), a type of the most lethal brain tumor, has remained a formidable challenge in terms of its treatment. Recently, many novel discoveries have underlined the regulatory roles of neurotransmitters in the microenvironment both physiologically and pathologically. By targeting the receptors synaptically or non-synaptically, neurotransmitters activate multiple signaling pathways. Significantly, many ligands acting on neurotransmitter receptors have shown great potential for inhibiting GBM growth and development, requiring further research. Here, we provide an overview of the most novel advances concerning the role of neurotransmitters in the normal neural and the GBM microenvironments, and discuss potential targeted drugs used for GBM treatment.
Collapse
|
21
|
Albertini C, Salerno A, Atzeni S, Uliassi E, Massenzio F, Maruca A, Rocca R, Mecava M, Silva FSG, Mena D, Valente P, Duarte AI, Chavarria D, Bissaro M, Moro S, Federico S, Spalluto G, Soukup O, Borges F, Alcaro S, Monti B, Oliveira PJ, Menéndez JC, Bolognesi ML. Riluzole-Rasagiline Hybrids: Toward the Development of Multi-Target-Directed Ligands for Amyotrophic Lateral Sclerosis. ACS Chem Neurosci 2022; 13:2252-2260. [PMID: 35868251 PMCID: PMC9354084 DOI: 10.1021/acschemneuro.2c00261] [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] [Indexed: 11/29/2022] Open
Abstract
![]()
Polypharmacology is a new trend in amyotrophic lateral
sclerosis
(ALS) therapy and an effective way of addressing a multifactorial
etiology involving excitotoxicity, mitochondrial dysfunction, oxidative
stress, and microglial activation. Inspired by a reported clinical
trial, we converted a riluzole (1)–rasagiline
(2) combination into single-molecule multi-target-directed
ligands. By a ligand-based approach, the highly structurally integrated
hybrids 3–8 were designed and synthesized.
Through a target- and phenotypic-based screening pipeline, we identified
hit compound 6. It showed monoamine oxidase A (MAO-A)
inhibitory activity (IC50 = 6.9 μM) rationalized
by in silico studies as well as in vitro brain permeability. By using neuronal and non-neuronal cell models,
including ALS-patient-derived cells, we disclosed for 6 a neuroprotective/neuroinflammatory profile similar to that of the
parent compounds and their combination. Furthermore, the unexpected
MAO inhibitory activity of 1 (IC50 = 8.7 μM)
might add a piece to the puzzle of its anti-ALS molecular profile.
Collapse
Affiliation(s)
- Claudia Albertini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Alessandra Salerno
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Silvia Atzeni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Francesca Massenzio
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Annalisa Maruca
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy.,Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
| | - Roberta Rocca
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy.,Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
| | - Marko Mecava
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Králové, Czech Republic
| | - Filomena S G Silva
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal.,Mitotag Lda, Biocant Park, 3060-197 Cantanhede, Portugal
| | - Débora Mena
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Pedro Valente
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal.,Research Unit for Sport and Physical Activity (CIDAF), Faculty of Sport Science and Physical Education (FCDEF-UC), University of Coimbra, 3040-248 Coimbra, Portugal
| | - Ana I Duarte
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Daniel Chavarria
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Maicol Bissaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Stefano Moro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Stephanie Federico
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giampiero Spalluto
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
| | - Ondřej Soukup
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Králové, Czech Republic
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy.,Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Paulo J Oliveira
- Center for Neuroscience and Cell Biology (CNC), Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Josè C Menéndez
- Department of Chemistry in Pharmaceutical Sciences, Organic and Medicinal Chemistry Unit, Faculty of Pharmacy, Universidad Complutense, 28040 Madrid, Spain
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
22
|
Liu TW, Chen CM, Chang KH. Biomarker of Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2022; 23:ijms23084148. [PMID: 35456966 PMCID: PMC9028544 DOI: 10.3390/ijms23084148] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
Parkinson's disease (PD) is caused by abnormal accumulation of α-synuclein in dopaminergic neurons of the substantia nigra, which subsequently causes motor symptoms. Neuroinflammation plays a vital role in the pathogenesis of neurodegeneration in PD. This neuroinflammatory neurodegeneration involves the activation of microglia, upregulation of proinflammatory factors, and gut microbiota. In this review, we summarized the recent findings on detection of PD by using inflammatory biomarkers, such as interleukin (IL)-1β, IL-2, IL-6, IL-10, tumor necrosis factor (TNF)-α; regulated upon activation, normal T cell expressed and presumably secreted (RANTES) and high-sensitivity c-reactive protein (hsCRP); and radiotracers such as [11C]PK11195 and [18F]-FEPPA, as well as by monitoring disease progression and the treatment response. Many PD-causing mutations in SNCA, LRRK2, PRKN, PINK1, and DJ-1 are also associated with neuroinflammation. Several anti-inflammatory medications, including nonsteroidal anti-inflammatory drugs (NSAID), inhibitors of TNF-α and NLR family pyrin domain containing 3 (NLRP3), agonists of nuclear factor erythroid 2-related factor 2 (NRF2), peroxisome proliferator-activated receptor gamma (PPAR-γ), and steroids, have demonstrated neuroprotective effects in in vivo or in vitro PD models. Clinical trials applying objective biomarkers are required to investigate the therapeutic potential of anti-inflammatory medications for PD.
Collapse
Affiliation(s)
- Tsai-Wei Liu
- Linkou Medical Center, Department of Neurology, Chang Gung Memorial Hospital, Tauoyan 333, Taiwan; (T.-W.L.); (C.-M.C.)
| | - Chiung-Mei Chen
- Linkou Medical Center, Department of Neurology, Chang Gung Memorial Hospital, Tauoyan 333, Taiwan; (T.-W.L.); (C.-M.C.)
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Kuo-Hsuan Chang
- Linkou Medical Center, Department of Neurology, Chang Gung Memorial Hospital, Tauoyan 333, Taiwan; (T.-W.L.); (C.-M.C.)
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Correspondence: ; Tel.: +886-3-3281200 (ext. 8729); Fax: +886-3-3288849
| |
Collapse
|
23
|
Zhao N, Li Y, Wang C, Xue Y, Peng L, Wang T, Zhao Y, Xu G, Yu S. DJ-1 activates the Atg5-Atg12-Atg16L1 complex via Sirt1 to influence microglial polarization and alleviate cerebral ischemia/reperfusion-induced inflammatory injury. Neurochem Int 2022; 157:105341. [PMID: 35429577 DOI: 10.1016/j.neuint.2022.105341] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 12/22/2022]
|
24
|
Magistrelli L, Contaldi E, Vignaroli F, Gallo S, Colombatto F, Cantello R, Comi C. Immune Response Modifications in the Genetic Forms of Parkinson's Disease: What Do We Know? Int J Mol Sci 2022; 23:ijms23073476. [PMID: 35408836 PMCID: PMC8998358 DOI: 10.3390/ijms23073476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized by loss of dopaminergic neurons in the pars compacta of the midbrain substantia nigra. PD pathophysiology is complex, multifactorial, and not fully understood yet. Nonetheless, recent data show that immune system hyperactivation with concomitant production of pro-inflammatory cytokines, both in the central nervous system (CNS) and the periphery, is a signature of idiopathic PD. About 5% of PD patients present an early onset with a determined genetic cause, with either autosomal dominant or recessive inheritance. The involvement of immunity in the genetic forms of PD has been a matter of interest in several recent studies. In this review, we will summarize the main findings of this new and promising field of research.
Collapse
Affiliation(s)
- Luca Magistrelli
- PhD Program in Clinical and Experimental Medicine and Medical Humanities, University of Insubria, 21100 Varese, Italy;
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.C.); (F.V.); (S.G.); (F.C.); (R.C.)
| | - Elena Contaldi
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.C.); (F.V.); (S.G.); (F.C.); (R.C.)
- PhD Program in Medical Sciences and Biotechnology, University of Piemonte Orientale, 28100 Novara, Italy
| | - Francesca Vignaroli
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.C.); (F.V.); (S.G.); (F.C.); (R.C.)
| | - Silvia Gallo
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.C.); (F.V.); (S.G.); (F.C.); (R.C.)
| | - Federico Colombatto
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.C.); (F.V.); (S.G.); (F.C.); (R.C.)
| | - Roberto Cantello
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.C.); (F.V.); (S.G.); (F.C.); (R.C.)
| | - Cristoforo Comi
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (E.C.); (F.V.); (S.G.); (F.C.); (R.C.)
- Correspondence:
| |
Collapse
|
25
|
Un H, Ugan RA, Kose D, Yayla M, Tastan TB, Bayir Y, Halici Z. A new approach to sepsis treatment by rasagiline: a molecular, biochemical and histopathological study. Mol Biol Rep 2022; 49:3875-3883. [PMID: 35301652 DOI: 10.1007/s11033-022-07235-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 01/05/2022] [Accepted: 02/07/2022] [Indexed: 11/25/2022]
Abstract
AIM We aimed to investigate the effects of rasagiline on acute lung injury that develops in the sepsis model induced with the cecal ligation and puncture in rats. MAIN METHODS The rats were separated into the following six groups, Group 1: Sham, Group 2: Sham + Rasagiline 4 mg/kg, Group 3: Sepsis, Group 4: Sepsis + Rasagiline 1 mg/kg, Group 5: Sepsis + Rasagiline 2 mg/kg, Group 6: Sepsis + Rasagiline 4 mg/kg. A total of four holes were opened with a 16-gauge needle through the cecum distal to the point of ligation. KEY FINDINGS Rasagiline treatment increased glutathione level and superoxide dismutase activity while decreased the malondialdehyde level after the sepsis. There was a statistically significant improvement in the doses of 2 mg/kg and 4 mg/kg. Rasagiline also increased Tnf-α, IL1β, IL6, NF-κβand HMGB1 gene expressions in dose-dependent at 2 mg/kg and 4 mg/kg doses. In increased doses, rasagiline prevent the development of edema, the formation of inflammation, and hemorrhage. SIGNIFICANCE Rasagiline exerts both antioxidant and anti-inflammatory effects on the cecal ligation and puncture induced acute lung injury in rats.
Collapse
Affiliation(s)
- Harun Un
- Faculty of Pharmacy, Department of Biochemistry, Agri Ibrahim Cecen University, Agri, Turkey.
| | - Rustem Anil Ugan
- Faculty of Pharmacy, Department of Pharmacology, Ataturk University, Erzurum, Turkey
| | - Duygu Kose
- Faculty of Medicine, Department of Pharmacology, Ataturk University, Erzurum, Turkey.,Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| | - Muhammed Yayla
- Faculty of Medicine, Department of Pharmacology, Kafkas University, Kars, Turkey
| | - Tugba Bal Tastan
- Faculty of Medicine, Department of Histology and Embryology, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Yasin Bayir
- Faculty of Pharmacy, Department of Biochemistry, Ataturk University, Erzurum, Turkey
| | - Zekai Halici
- Faculty of Medicine, Department of Pharmacology, Ataturk University, Erzurum, Turkey.,Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| |
Collapse
|
26
|
Li XX, Zhang F. Targeting TREM2 for Parkinson's Disease: Where to Go? Front Immunol 2022; 12:795036. [PMID: 35003116 PMCID: PMC8740229 DOI: 10.3389/fimmu.2021.795036] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/13/2021] [Indexed: 12/16/2022] Open
Abstract
Parkinson’s disease (PD) is one of most common neurodegenerative disorders caused by a combination of environmental and genetic risk factors. Currently, numerous population genetic studies have shown that polymorphisms in myeloid cell-triggered receptor II (TREM2) are associated with a variety of neurodegenerative disorders. Recently, TREM2 has been verified to represent a promising candidate gene for PD susceptibility and progression. For example, the expression of TREM2 was apparently increased in the prefrontal cortex of PD patients. Moreover, the rare missense mutations in TREM2 (rs75932628, p.R47H) was confirmed to be a risk factor of PD. In addition, overexpression of TREM2 reduced dopaminergic neurodegeneration in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine mouse model of PD. Due to the complex pathogenesis of PD, there is still no effective drug treatment. Thus, TREM2 has received increasing widespread attention as a potential therapeutic target. This review focused on the variation of TREM2 in PD and roles of TREM2 in PD pathogenesis, such as excessive-immune inflammatory response, α-Synuclein aggregation and oxidative stress, to further provide evidence for new immune-related biomarkers and therapies for PD.
Collapse
Affiliation(s)
- Xiao-Xian Li
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Feng Zhang
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China.,Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| |
Collapse
|
27
|
Putnins EE, Goebeler V, Ostadkarampour M. Monoamine Oxidase-B Inhibitor Reduction in Pro-Inflammatory Cytokines Mediated by Inhibition of cAMP-PKA/EPAC Signaling. Front Pharmacol 2021; 12:741460. [PMID: 34867348 PMCID: PMC8635787 DOI: 10.3389/fphar.2021.741460] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
Mucosal epithelial cell integrity is an important component of innate immunity and it protects the host from an environment rich in microorganisms. Virulence factors from Gram-negative bacteria [e.g. lipopolysaccharide (LPS)] induce significant pro-inflammatory cytokine expression. Monoamine oxidase (MAO) inhibitors reduce cytokine expression in a variety of inflammatory models and may therefore have therapeutic potential for a number of inflammatory diseases. We tested the anti-inflammatory therapeutic potential of a recently developed reversible MAO-B inhibitor (RG0216) with reduced transport across the blood–brain barrier. In an epithelial cell culture model, RG0216 significantly decreased LPS-induced interleukin (IL)-6 and IL-1β gene and protein expression and was as effective as equimolar concentrations of deprenyl (an existing irreversible MAO-B inhibitor). Hydrogen peroxide and modulating dopamine receptor signaling had no effect on cytokine expression. We showed that LPS-induced expression of IL-6 and IL-1β was cAMP dependent, that IL-6 and IL-1β expression were induced by direct cAMP activation (forskolin) and that RG0216 and deprenyl effectively reduced cAMP-mediated cytokine expression. Targeted protein kinase A (PKA) and Exchange Protein Activated by cAMP (EPAC) activation regulated IL-6 and IL-1β expression, albeit in different ways, but both cytokines were effectively decreased with RG0216. RG0216 reduction of LPS-induced cytokine expression occurred by acting downstream of the cAMP-PKA/EPAC signaling cascade. This represents a novel mechanism by which MAO-B selective inhibitors regulate LPS-induced IL-6 and IL-1β expression.
Collapse
Affiliation(s)
- Edward E Putnins
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, BC, Canada
| | - Verena Goebeler
- Department of Pediatrics, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Mahyar Ostadkarampour
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
28
|
DJ-1 inhibits microglial activation and protects dopaminergic neurons in vitro and in vivo through interacting with microglial p65. Cell Death Dis 2021; 12:715. [PMID: 34274951 PMCID: PMC8286256 DOI: 10.1038/s41419-021-04002-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD), one of the most common neurodegenerative disorders, is characterized by progressive neurodegeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). DJ-1 acts essential roles in neuronal protection and anti-neuroinflammatory response, and its loss of function is tightly associated with a familial recessive form of PD. However, the molecular mechanism of DJ-1 involved in neuroinflammation is largely unclear. Here, we found that wild-type DJ-1, rather than the pathogenic L166P mutant DJ-1, directly binds to the subunit p65 of nuclear factor-κB (NF-κB) in the cytoplasm, and loss of DJ-1 promotes p65 nuclear translocation by facilitating the dissociation between p65 and NF-κB inhibitor α (IκBα). DJ-1 knockout (DJ-1-/-) mice exhibit more microglial activation compared with wild-type littermate controls, especially in response to lipopolysaccharide (LPS) treatment. In cellular models, knockdown of DJ-1 significantly upregulates the gene expression and increases the release of LPS-treated inflammatory cytokines in primary microglia and BV2 cells. Furthermore, DJ-1 deficiency in microglia significantly enhances the neuronal toxicity in response to LPS stimulus. In addition, pharmacological blockage of NF-κB nuclear translocation by SN-50 prevents microglial activation and alleviates the damage of DA neurons induced by microglial DJ-1 deficiency in vivo and in vitro. Thus, our data illustrate a novel mechanism by which DJ-1 facilitates the interaction between IκBα and p65 by binding to p65 in microglia, and thus repressing microglial activation and exhibiting the protection of DA neurons from neuroinflammation-mediated injury in PD.
Collapse
|
29
|
Huang M, Chen S. DJ-1 in neurodegenerative diseases: Pathogenesis and clinical application. Prog Neurobiol 2021; 204:102114. [PMID: 34174373 DOI: 10.1016/j.pneurobio.2021.102114] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/22/2021] [Accepted: 06/21/2021] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases (NDs) are one of the major health threats to human characterized by selective and progressive neuronal loss. The mechanisms of NDs are still not fully understood. The study of genetic defects and disease-related proteins offers us a window into the mystery of it, and the extension of knowledge indicates that different NDs share similar features, mechanisms, and even genetic or protein abnormalities. Among these findings, PARK7 and its production DJ-1 protein, which was initially found implicated in PD, have also been found altered in other NDs. PARK7 mutations, altered expression and posttranslational modification (PTM) cause DJ-1 abnormalities, which in turn lead to downstream mechanisms shared by most NDs, such as mitochondrial dysfunction, oxidative stress, protein aggregation, autophagy defects, and so on. The knowledge of DJ-1 derived from PD researches might apply to other NDs in both basic research and clinical application, and might yield novel insights into and alternative approaches for dealing with NDs.
Collapse
Affiliation(s)
- Maoxin Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China; Lab for Translational Research of Neurodegenerative Diseases, Institute of Immunochemistry, Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
30
|
Magnusen AF, Hatton SL, Rani R, Pandey MK. Genetic Defects and Pro-inflammatory Cytokines in Parkinson's Disease. Front Neurol 2021; 12:636139. [PMID: 34239490 PMCID: PMC8259624 DOI: 10.3389/fneur.2021.636139] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is a movement disorder attributed to the loss of dopaminergic (DA) neurons mainly in the substantia nigra pars compacta. Motor symptoms include resting tremor, rigidity, and bradykinesias, while non-motor symptoms include autonomic dysfunction, anxiety, and sleeping problems. Genetic mutations in a number of genes (e.g., LRRK2, GBA, SNCA, PARK2, PARK6, and PARK7) and the resultant abnormal activation of microglial cells are assumed to be the main reasons for the loss of DA neurons in PD with genetic causes. Additionally, immune cell infiltration and their participation in major histocompatibility complex I (MHCI) and/or MHCII-mediated processing and presentation of cytosolic or mitochondrial antigens activate the microglial cells and cause the massive generation of pro-inflammatory cytokines and chemokines, which are all critical for the propagation of brain inflammation and the neurodegeneration in PD with genetic and idiopathic causes. Despite knowing the involvement of several of such immune devices that trigger neuroinflammation and neurodegeneration in PD, the exact disease mechanism or the innovative biomarker that could detect disease severity in PD linked to LRRK2, GBA, SNCA, PARK2, PARK6, and PARK7 defects is largely unknown. The current review has explored data from genetics, immunology, and in vivo and ex vivo functional studies that demonstrate that certain genetic defects might contribute to microglial cell activation and massive generation of a number of pro-inflammatory cytokines and chemokines, which ultimately drive the brain inflammation and lead to neurodegeneration in PD. Understanding the detailed involvement of a variety of immune mediators, their source, and the target could provide a better understanding of the disease process. This information might be helpful in clinical diagnosis, monitoring of disease progression, and early identification of affected individuals.
Collapse
Affiliation(s)
- Albert Frank Magnusen
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Shelby Loraine Hatton
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Reena Rani
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Manoj Kumar Pandey
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Department of Paediatrics of University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
31
|
MacMahon Copas AN, McComish SF, Fletcher JM, Caldwell MA. The Pathogenesis of Parkinson's Disease: A Complex Interplay Between Astrocytes, Microglia, and T Lymphocytes? Front Neurol 2021; 12:666737. [PMID: 34122308 PMCID: PMC8189423 DOI: 10.3389/fneur.2021.666737] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disease, is characterised by the motor symptoms of bradykinesia, rigidity and resting tremor and non-motor symptoms of sleep disturbances, constipation, and depression. Pathological hallmarks include neuroinflammation, degeneration of dopaminergic neurons in the substantia nigra pars compacta, and accumulation of misfolded α-synuclein proteins as intra-cytoplasmic Lewy bodies and neurites. Microglia and astrocytes are essential to maintaining homeostasis within the central nervous system (CNS), including providing protection through the process of gliosis. However, dysregulation of glial cells results in disruption of homeostasis leading to a chronic pro-inflammatory, deleterious environment, implicated in numerous CNS diseases. Recent evidence has demonstrated a role for peripheral immune cells, in particular T lymphocytes in the pathogenesis of PD. These cells infiltrate the CNS, and accumulate in the substantia nigra, where they secrete pro-inflammatory cytokines, stimulate surrounding immune cells, and induce dopaminergic neuronal cell death. Indeed, a greater understanding of the integrated network of communication that exists between glial cells and peripheral immune cells may increase our understanding of disease pathogenesis and hence provide novel therapeutic approaches.
Collapse
Affiliation(s)
- Adina N MacMahon Copas
- Department of Physiology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sarah F McComish
- Department of Physiology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Jean M Fletcher
- Department of Physiology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland.,School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Maeve A Caldwell
- Department of Physiology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
32
|
Assessing the Anti-Inflammatory Activity of the Anxiolytic Drug Buspirone Using CRISPR-Cas9 Gene Editing in LPS-Stimulated BV-2 Microglial Cells. Cells 2021; 10:cells10061312. [PMID: 34070429 PMCID: PMC8229595 DOI: 10.3390/cells10061312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 01/06/2023] Open
Abstract
Buspirone is an anxiolytic drug with robust serotonin receptor 1A (Htr1a) agonist activities. However, evidence has demonstrated that this drug also targets the dopamine D3 receptor (Drd3), where it acts as a potent antagonist. In vivo, Drd3 blockade is neuroprotective and reduces inflammation in models of Parkinson’s disease. To test if buspirone also elicited anti-inflammatory activities in vitro, we generated stable Drd3−/− and Htr1a−/− BV2 microglial cell lines using CRISPR-Cas9 technology and then tested the effects of buspirone after lipopolysaccharide (LPS) challenge. We found that LPS exposure had no effect on cell viability, except in Htr1a−/− cells, where viability was reduced (p < 0.001). Drug treatment reduced viability in Drd3−/− cells, but not in WT or Htr1a−/− cells. Buspirone counteracted LPS-induced NO release, NOS2, IL-1β and TNF-α gene expression in WT cells, whereas it exerted limited effects in Drd3−/− or Htr1a−/− microglia. In summary, our findings indicate that buspirone attenuates microglial polarization after LPS challenge. These results also highlight some major effects of Drd3 or Htr1a genetic ablation on microglial biology, raising important questions on the complex role of neurotransmitters in regulating microglia functions.
Collapse
|
33
|
Ostadkarampour M, Putnins EE. Monoamine Oxidase Inhibitors: A Review of Their Anti-Inflammatory Therapeutic Potential and Mechanisms of Action. Front Pharmacol 2021; 12:676239. [PMID: 33995107 PMCID: PMC8120032 DOI: 10.3389/fphar.2021.676239] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/06/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic inflammatory diseases are debilitating, affect patients' quality of life, and are a significant financial burden on health care. Inflammation is regulated by pro-inflammatory cytokines and chemokines that are expressed by immune and non-immune cells, and their expression is highly controlled, both spatially and temporally. Their dysregulation is a hallmark of chronic inflammatory and autoimmune diseases. Significant evidence supports that monoamine oxidase (MAO) inhibitor drugs have anti-inflammatory effects. MAO inhibitors are principally prescribed for the management of a variety of central nervous system (CNS)-associated diseases such as depression, Alzheimer's, and Parkinson's; however, they also have anti-inflammatory effects in the CNS and a variety of non-CNS tissues. To bolster support for their development as anti-inflammatories, it is critical to elucidate their mechanism(s) of action. MAO inhibitors decrease the generation of end products such as hydrogen peroxide, aldehyde, and ammonium. They also inhibit biogenic amine degradation, and this increases cellular and pericellular catecholamines in a variety of immune and some non-immune cells. This decrease in end product metabolites and increase in catecholamines can play a significant role in the anti-inflammatory effects of MAO inhibitors. This review examines MAO inhibitor effects on inflammation in a variety of in vitro and in vivo CNS and non-CNS disease models, as well as their anti-inflammatory mechanism(s) of action.
Collapse
Affiliation(s)
- Mahyar Ostadkarampour
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, BC, Canada
| | - Edward E Putnins
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
34
|
Badanjak K, Fixemer S, Smajić S, Skupin A, Grünewald A. The Contribution of Microglia to Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2021; 22:4676. [PMID: 33925154 PMCID: PMC8125756 DOI: 10.3390/ijms22094676] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/19/2021] [Accepted: 04/24/2021] [Indexed: 12/12/2022] Open
Abstract
With the world's population ageing, the incidence of Parkinson's disease (PD) is on the rise. In recent years, inflammatory processes have emerged as prominent contributors to the pathology of PD. There is great evidence that microglia have a significant neuroprotective role, and that impaired and over activated microglial phenotypes are present in brains of PD patients. Thereby, PD progression is potentially driven by a vicious cycle between dying neurons and microglia through the instigation of oxidative stress, mitophagy and autophagy dysfunctions, a-synuclein accumulation, and pro-inflammatory cytokine release. Hence, investigating the involvement of microglia is of great importance for future research and treatment of PD. The purpose of this review is to highlight recent findings concerning the microglia-neuronal interplay in PD with a focus on human postmortem immunohistochemistry and single-cell studies, their relation to animal and iPSC-derived models, newly emerging technologies, and the resulting potential of new anti-inflammatory therapies for PD.
Collapse
Affiliation(s)
- Katja Badanjak
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Esch-sur-Alzette, Luxembourg; (K.B.); (S.F.); (S.S.); (A.S.)
| | - Sonja Fixemer
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Esch-sur-Alzette, Luxembourg; (K.B.); (S.F.); (S.S.); (A.S.)
- Luxembourg Centre for Neuropathology (LCNP), L-3555 Dudelange, Luxembourg
| | - Semra Smajić
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Esch-sur-Alzette, Luxembourg; (K.B.); (S.F.); (S.S.); (A.S.)
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Esch-sur-Alzette, Luxembourg; (K.B.); (S.F.); (S.S.); (A.S.)
- Department of Neuroscience, University California San Diego, La Jolla, CA 92093, USA
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Esch-sur-Alzette, Luxembourg; (K.B.); (S.F.); (S.S.); (A.S.)
- Institute of Neurogenetics, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
35
|
Pons-Espinal M, Blasco-Agell L, Consiglio A. Dissecting the non-neuronal cell contribution to Parkinson's disease pathogenesis using induced pluripotent stem cells. Cell Mol Life Sci 2021; 78:2081-2094. [PMID: 33210214 PMCID: PMC7966189 DOI: 10.1007/s00018-020-03700-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 10/10/2020] [Accepted: 10/31/2020] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD) is an incurable age-linked neurodegenerative disease with characteristic movement impairments that are caused by the progressive loss of dopamine-containing neurons (DAn) within the substantia nigra pars compacta. It has been suggested that misfolded protein aggregates together with neuroinflammation and glial reactivity, may impact nerve cell function, leading to neurodegeneration and diseases, such as PD. However, not many studies have been able to examine the role of human glial cells in the pathogenesis of PD. With the advent of induced pluripotent stem cell (iPSC) technology, it is now possible to reprogram human somatic cells to pluripotency and to generate viable human patient-specific DA neurons and glial cells, providing a tremendous opportunity for dissecting cellular and molecular pathological mechanisms occurring at early stages of PD. This reviews will report on recent work using human iPSC and 3D brain organoid models showing that iPSC technology can be used to recapitulate PD-relevant disease-associated phenotypes, including protein aggregation, cell death or loss of neurite complexity and deficient autophagic vacuoles clearance and focus on the recent co-culture systems that are revealing new insights into the complex interactions that occur between different brain cell types during neurodegeneration. Consequently, such advances are the key to improve our understanding of PD pathology and generate potential targets for new therapies aimed at curing PD patients.
Collapse
Affiliation(s)
- Meritxell Pons-Espinal
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, 08908, Hospitalet de Llobregat, Spain.
- Institute of Biomedicine (IBUB) of the University of Barcelona (UB), 08028, Barcelona, Spain.
| | - Lucas Blasco-Agell
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, 08908, Hospitalet de Llobregat, Spain
- Institute of Biomedicine (IBUB) of the University of Barcelona (UB), 08028, Barcelona, Spain
| | - Antonella Consiglio
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, 08908, Hospitalet de Llobregat, Spain.
- Institute of Biomedicine (IBUB) of the University of Barcelona (UB), 08028, Barcelona, Spain.
- Department of Molecular and Translational Medicine, University of Brescia, Piazza del Mercato, 15, 25121, Brescia, BS, Italy.
| |
Collapse
|
36
|
Sivasubramaniyam T, Yang J, Cheng HS, Zyla A, Li A, Besla R, Dotan I, Revelo XS, Shi SY, Le H, Schroer SA, Dodington DW, Park YJ, Kim MJ, Febbraro D, Ruel I, Genest J, Kim RH, Mak TW, Winer DA, Robbins CS, Woo M. Dj1 deficiency protects against atherosclerosis with anti-inflammatory response in macrophages. Sci Rep 2021; 11:4723. [PMID: 33633277 PMCID: PMC7907332 DOI: 10.1038/s41598-021-84063-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/08/2021] [Indexed: 11/09/2022] Open
Abstract
Inflammation is a key contributor to atherosclerosis with macrophages playing a pivotal role through the induction of oxidative stress and cytokine/chemokine secretion. DJ1, an anti-oxidant protein, has shown to paradoxically protect against chronic and acute inflammation. However, the role of DJ1 in atherosclerosis remains elusive. To assess the role of Dj1 in atherogenesis, we generated whole-body Dj1-deficient atherosclerosis-prone Apoe null mice (Dj1-/-Apoe-/-). After 21 weeks of atherogenic diet, Dj1-/- Apoe-/-mice were protected against atherosclerosis with significantly reduced plaque macrophage content. To assess whether haematopoietic or parenchymal Dj1 contributed to atheroprotection in Dj1-deficient mice, we performed bone-marrow (BM) transplantation and show that Dj1-deficient BM contributed to their attenuation in atherosclerosis. To assess cell-autonomous role of macrophage Dj1 in atheroprotection, BM-derived macrophages from Dj1-deficient mice and Dj1-silenced macrophages were assessed in response to oxidized low-density lipoprotein (oxLDL). In both cases, there was an enhanced anti-inflammatory response which may have contributed to atheroprotection in Dj1-deficient mice. There was also an increased trend of plasma DJ-1 levels from individuals with ischemic heart disease compared to those without. Our findings indicate an atheropromoting role of Dj1 and suggests that targeting Dj1 may provide a novel therapeutic avenue for atherosclerosis treatment or prevention.
Collapse
Affiliation(s)
- Tharini Sivasubramaniyam
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Jiaqi Yang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Henry S Cheng
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Alexandra Zyla
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Angela Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Rickvinder Besla
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Idit Dotan
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Xavier S Revelo
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Sally Yu Shi
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Helen Le
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Stephanie A Schroer
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - David W Dodington
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Yoo Jin Park
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Min Jeong Kim
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181, Korea
| | - Daniella Febbraro
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Isabelle Ruel
- Research Institute of the McGill University Health Centre, Royal Victoria Hospital, Montreal, QC, H4A 3J1, Canada
| | - Jacques Genest
- Research Institute of the McGill University Health Centre, Royal Victoria Hospital, Montreal, QC, H4A 3J1, Canada.,Department of Medicine, McGill University, Royal Victoria Hospital, Montreal, QC, H4A 3J1, Canada
| | - Raymond H Kim
- Department of Medicine, University Health Network/Sinai Health System, University of Toronto, Toronto, ON, M5G 2C4, Canada
| | - Tak W Mak
- Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Daniel A Winer
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G 2M9, Canada.,Department of Pathology, University Health Network, Toronto, M5G 2C4, Canada
| | - Clinton S Robbins
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, M5G 2M9, Canada. .,Department of Immunology, University of Toronto, Toronto, ON, M5G 2M9, Canada. .,Department of Medicine, University Health Network/Sinai Health System, University of Toronto, Toronto, ON, M5G 2C4, Canada. .,Division of Endocrinology and Metabolism, University Health Network/Sinai Health System, University of Toronto, Toronto, ON, M5G 2C4, Canada. .,MaRS Centre, Toronto Medical Discovery Tower, 101 College Street, 10th floor, Room 10-361, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
37
|
Wang T, Zhao N, Peng L, Li Y, Huang X, Zhu J, Chen Y, Yu S, Zhao Y. DJ-1 Regulates Microglial Polarization Through P62-Mediated TRAF6/IRF5 Signaling in Cerebral Ischemia-Reperfusion. Front Cell Dev Biol 2020; 8:593890. [PMID: 33392187 PMCID: PMC7773790 DOI: 10.3389/fcell.2020.593890] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/25/2020] [Indexed: 12/28/2022] Open
Abstract
The polarization of microglia/macrophage, the resident immune cells in the brain, plays an important role in the injury and repair associated with ischemia-reperfusion (I/R). Previous studies have shown that DJ-1 has a protective effect in cerebral I/R. We found that DJ-1 regulates the polarization of microglial cells/macrophages after cerebral I/R and explored the mechanism by which DJ-1 mediates microglial/macrophage polarization in cerebral I/R. Middle cerebral artery occlusion/reperfusion (MCAO/R) and oxygen and glucose deprivation/reoxygenation (OGD/R) models were used to simulate cerebral I/R in vivo and in vitro, respectively. DJ-1 siRNA and the DJ-1-based polypeptide ND13 were used to produce an effect on DJ-1, and the P62-specific inhibitor XRK3F2 was used to block the effect of P62. Enhancing the expression of DJ-1 induced anti-inflammatory (M2) polarization of microglia/macrophage, and the expression of the anti-inflammatory factors IL-10 and IL-4 increased. Interference with DJ-1 expression induced pro-inflammatory (M1) polarization of microglia/macrophage, and the expression of the proinflammatory factors TNF-α and IL-1β increased. DJ-1 inhibited the expression of P62, impeded the interaction between P62 and TRAF6, and blocked nuclear entry of IRF5. In subsequent experiments, XRK3F2 synergistically promoted the effect of DJ-1 on microglial/macrophage polarization, further attenuating the interaction between P62 and TRAF6.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Na Zhao
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Li Peng
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yumei Li
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Xiaohuan Huang
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Jin Zhu
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yanlin Chen
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Shanshan Yu
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing, China.,Molecular Medical Laboratory, Chongqing Medical University, Chongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
38
|
Stolero N, Frenkel D. The dialog between neurons and microglia in Alzheimer's disease: The neurotransmitters view. J Neurochem 2020; 158:1412-1424. [PMID: 33314073 DOI: 10.1111/jnc.15262] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/12/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
Microglia play a vital role in maintaining brain homeostasis. Their continuous sensing of surrounding micro-environments is crucial for their activity. Cross talk between specific neurons and microglia might occur through specific neurotransmitter receptors on microglia. Impairment with this interaction might result in pathological activity of microglia against potential insults. The reason for this activity in many neurodegenerative diseases such as Alzheimer's disease (AD) is not known. However, several papers report of the effects of different neurotransmitter agonists on microglial cells function that relate to their activity in AD. This review aims to summarize those works and to raise potential fundamental questions for future research.
Collapse
Affiliation(s)
- Nofar Stolero
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
39
|
The Emerging Role of the Lysosome in Parkinson's Disease. Cells 2020; 9:cells9112399. [PMID: 33147750 PMCID: PMC7692401 DOI: 10.3390/cells9112399] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/22/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Lysosomal function has a central role in maintaining neuronal homeostasis, and, accordingly, lysosomal dysfunction has been linked to neurodegeneration and particularly to Parkinson’s disease (PD). Lysosomes are the converging step where the substrates delivered by autophagy and endocytosis are degraded in order to recycle their primary components to rebuild new macromolecules. Genetic studies have revealed the important link between the lysosomal function and PD; several of the autosomal dominant and recessive genes associated with PD as well as several genetic risk factors encode for lysosomal, autophagic, and endosomal proteins. Mutations in these PD-associated genes can cause lysosomal dysfunction, and since α-synuclein degradation is mostly lysosomal-dependent, among other consequences, lysosomal impairment can affect α-synuclein turnover, contributing to increase its intracellular levels and therefore promoting its accumulation and aggregation. Recent studies have also highlighted the bidirectional link between Parkinson’s disease and lysosomal storage diseases (LSD); evidence includes the presence of α-synuclein inclusions in the brain regions of patients with LSD and the identification of several lysosomal genes involved in LSD as genetic risk factors to develop PD.
Collapse
|
40
|
Kam TI, Hinkle JT, Dawson TM, Dawson VL. Microglia and astrocyte dysfunction in parkinson's disease. Neurobiol Dis 2020; 144:105028. [PMID: 32736085 PMCID: PMC7484088 DOI: 10.1016/j.nbd.2020.105028] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 07/01/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
While glia are essential for regulating the homeostasis in the normal brain, their dysfunction contributes to neurodegeneration in many brain diseases, including Parkinson's disease (PD). Recent studies have identified that PD-associated genes are expressed in glial cells as well as neurons and have crucial roles in microglia and astrocytes. Here, we discuss the role of microglia and astrocytes dysfunction in relation to PD-linked mutations and their implications in PD pathogenesis. A better understanding of microglia and astrocyte functions in PD may provide insights into neurodegeneration and novel therapeutic approaches for PD.
Collapse
Affiliation(s)
- Tae-In Kam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jared T Hinkle
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
41
|
Iron-responsive-like elements and neurodegenerative ferroptosis. ACTA ACUST UNITED AC 2020; 27:395-413. [PMID: 32817306 PMCID: PMC7433652 DOI: 10.1101/lm.052282.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022]
Abstract
A set of common-acting iron-responsive 5′untranslated region (5′UTR) motifs can fold into RNA stem loops that appear significant to the biology of cognitive declines of Parkinson's disease dementia (PDD), Lewy body dementia (LDD), and Alzheimer's disease (AD). Neurodegenerative diseases exhibit perturbations of iron homeostasis in defined brain subregions over characteristic time intervals of progression. While misfolding of Aβ from the amyloid-precursor-protein (APP), alpha-synuclein, prion protein (PrP) each cause neuropathic protein inclusions in the brain subregions, iron-responsive-like element (IRE-like) RNA stem–loops reside in their transcripts. APP and αsyn have a role in iron transport while gene duplications elevate the expression of their products to cause rare familial cases of AD and PDD. Of note, IRE-like sequences are responsive to excesses of brain iron in a potential feedback loop to accelerate neuronal ferroptosis and cognitive declines as well as amyloidosis. This pathogenic feedback is consistent with the translational control of the iron storage protein ferritin. We discuss how the IRE-like RNA motifs in the 5′UTRs of APP, alpha-synuclein and PrP mRNAs represent uniquely folded drug targets for therapies to prevent perturbed iron homeostasis that accelerates AD, PD, PD dementia (PDD) and Lewy body dementia, thus preventing cognitive deficits. Inhibition of alpha-synuclein translation is an option to block manganese toxicity associated with early childhood cognitive problems and manganism while Pb toxicity is epigenetically associated with attention deficit and later-stage AD. Pathologies of heavy metal toxicity centered on an embargo of iron export may be treated with activators of APP and ferritin and inhibitors of alpha-synuclein translation.
Collapse
|
42
|
Down-regulation of DJ-1 Augments Neuroinflammation via Nrf2/Trx1/NLRP3 Axis in MPTP-induced Parkinson's Disease Mouse Model. Neuroscience 2020; 442:253-263. [PMID: 32526245 DOI: 10.1016/j.neuroscience.2020.06.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/25/2022]
Abstract
Microglia-mediated neuroinflammation plays a significant role in the pathogenesis of Parkinson's disease (PD). Down-regulation of DJ-1, a PD-associated protein, has been recently found to increase microglial sensitivity to lipopolysaccharides (LPS). However, the role of DJ-1 in microglia-mediated neuroinflammation in PD remains unclear. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was used to establish a PD model with mice and tyrosine hydroxylase (TH) staining was performed to validate the model. Adenovirus strategy and shRNA was employed to knockdown the expression of DJ-1 in mice and BV2 microglia, respectively. Western Blot and quantitative PCR were carried out to determine the expression of cytokines, DJ-1, Nrf2, Trx1 and NRLP3. Immunoprecipitation was used to examine the potential interaction between DJ-1 and Nrf2 or Trx1. Flow cytometry-based Annexin V/7-AAD assay were performed to evaluate cell apoptosis. We found that down-regulation of DJ-1 exacerbated neuroinflammation in PD mice. DJ-1 and Nrf2 knockdown promoted inflammation and cell apoptosis in BV2 microglia, while NLRP3 knockdown had opposite effects. Furthermore, DJ-1 regulated the expression of NLRP3 by upregulating Nrf2/Trx1 axis. Taken together, these data suggested that down-regulation of DJ-1 accelerated microglia-mediated neuroinflammation and cell apoptosis via Nrf2/Trx1/NLRP3 axis. Thus, our results demonstrated the important role of DJ-1 in PD pathogenesis and warranted further investigation of DJ-1 as a therapeutic target for PD.
Collapse
|
43
|
Belloli S, Morari M, Murtaj V, Valtorta S, Moresco RM, Gilardi MC. Translation Imaging in Parkinson's Disease: Focus on Neuroinflammation. Front Aging Neurosci 2020; 12:152. [PMID: 32581765 PMCID: PMC7289967 DOI: 10.3389/fnagi.2020.00152] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the appearance of α-synuclein insoluble aggregates known as Lewy bodies. Neurodegeneration is accompanied by neuroinflammation mediated by cytokines and chemokines produced by the activated microglia. Several studies demonstrated that such an inflammatory process is an early event, and contributes to oxidative stress and mitochondrial dysfunctions. α-synuclein fibrillization and aggregation activate microglia and contribute to disease onset and progression. Mutations in different genes exacerbate the inflammatory phenotype in the monogenic compared to sporadic forms of PD. Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) with selected radiopharmaceuticals allow in vivo imaging of molecular modifications in the brain of living subjects. Several publications showed a reduction of dopaminergic terminals and dopamine (DA) content in the basal ganglia, starting from the early stages of the disease. Moreover, non-dopaminergic neuronal pathways are also affected, as shown by in vivo studies with serotonergic and glutamatergic radiotracers. The role played by the immune system during illness progression could be investigated with PET ligands that target the microglia/macrophage Translocator protein (TSPO) receptor. These agents have been used in PD patients and rodent models, although often without attempting correlations with other molecular or functional parameters. For example, neurodegeneration and brain plasticity can be monitored using the metabolic marker 2-Deoxy-2-[18F]fluoroglucose ([18F]-FDG), while oxidative stress can be probed using the copper-labeled diacetyl-bis(N-methyl-thiosemicarbazone) ([Cu]-ATSM) radioligand, whose striatal-specific binding ratio in PD patients seems to correlate with a disease rating scale and motor scores. Also, structural and functional modifications during disease progression may be evaluated by Magnetic Resonance Imaging (MRI), using different parameters as iron content or cerebral volume. In this review article, we propose an overview of in vivo clinical and non-clinical imaging research on neuroinflammation as an emerging marker of early PD. We also discuss how multimodal-imaging approaches could provide more insights into the role of the inflammatory process and related events in PD development.
Collapse
Affiliation(s)
- Sara Belloli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Michele Morari
- Section of Pharmacology, Department of Medical Sciences, National Institute for Neuroscience, University of Ferrara, Ferrara, Italy
| | - Valentina Murtaj
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Silvia Valtorta
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Rosa Maria Moresco
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Maria Carla Gilardi
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
44
|
Maron R, Armony G, Tsoory M, Wilchek M, Frenkel D, Arnon R. Peptide Interference with APP and Tau Association: Relevance to Alzheimer's Disease Amelioration. Int J Mol Sci 2020; 21:E3270. [PMID: 32380752 PMCID: PMC7246762 DOI: 10.3390/ijms21093270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 01/02/2023] Open
Abstract
The two major proteins involved in Alzheimer's disease (AD) are the amyloid precursor protein (APP) and Tau. Here, we demonstrate that these two proteins can bind to each other. Four possible peptides APP1 (390-412), APP2 (713-730), Tau1 (19-34) and Tau2 (331-348), were predicted to be involved in this interaction, with actual binding confirmed for APP1 and Tau1. In vivo studies were performed in an Alzheimer Disease animal model-APP double transgenic (Tg) 5xFAD-as well as in 5xFAD crossed with Tau transgenic 5xFADXTau (FT), which exhibit declined cognitive reduction at four months of age. Nasal administration of APP1 and Tau1 mixture, three times a week for four or five months, reduced amyloid plaque burden as well as the level of soluble Aβ 1-42 in the brain. The treatment prevented the deterioration of cognitive functions when initiated at the age of three months, before cognitive deficiency was evident, and also at the age of six months, when such deficiencies are already observed, leading to a full regain of cognitive function.
Collapse
Affiliation(s)
- Ruth Maron
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Gad Armony
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Meir Wilchek
- Department of Biomolecular Science, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Sagol School of Neuroscience Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Ruth Arnon
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
45
|
Miller YI, Navia-Pelaez JM, Corr M, Yaksh TL. Lipid rafts in glial cells: role in neuroinflammation and pain processing. J Lipid Res 2020; 61:655-666. [PMID: 31862695 PMCID: PMC7193960 DOI: 10.1194/jlr.tr119000468] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/06/2019] [Indexed: 12/27/2022] Open
Abstract
Activation of microglia and astrocytes secondary to inflammatory processes contributes to the development and perpetuation of pain with a neuropathic phenotype. This pain state presents as a chronic debilitating condition and affects a large population of patients with conditions like rheumatoid arthritis and diabetes, or after surgery, trauma, or chemotherapy. Here, we review the regulation of lipid rafts in glial cells and the role they play as a key component of neuroinflammatory sensitization of central pain signaling pathways. In this context, we introduce the concept of an inflammaraft (i-raft), enlarged lipid rafts harboring activated receptors and adaptor molecules and serving as an organizing platform to initiate inflammatory signaling and the cellular response. Characteristics of the inflammaraft include increased relative abundance of lipid rafts in inflammatory cells, increased content of cholesterol per raft, and increased levels of inflammatory receptors, such as toll-like receptor (TLR)4, adaptor molecules, ion channels, and enzymes in lipid rafts. This inflammaraft motif serves an important role in the membrane assembly of protein complexes, for example, TLR4 dimerization. Operating within this framework, we demonstrate the involvement of inflammatory receptors, redox molecules, and ion channels in the inflammaraft formation and the regulation of cholesterol and sphingolipid metabolism in the inflammaraft maintenance and disruption. Strategies for targeting inflammarafts, without affecting the integrity of lipid rafts in noninflammatory cells, may lead to developing novel therapies for neuropathic pain states and other neuroinflammatory conditions.
Collapse
Affiliation(s)
- Yury I Miller
- Departments of MedicineUniversity of California San Diego, La Jolla, CA. mailto:
| | | | - Maripat Corr
- Departments of MedicineUniversity of California San Diego, La Jolla, CA
| | - Tony L Yaksh
- Anesthesiology,University of California San Diego, La Jolla, CA
| |
Collapse
|
46
|
Lazdon E, Stolero N, Frenkel D. Microglia and Parkinson's disease: footprints to pathology. J Neural Transm (Vienna) 2020; 127:149-158. [DOI: 10.1007/s00702-020-02154-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/26/2020] [Indexed: 12/11/2022]
|
47
|
Szechtman H, Harvey BH, Woody EZ, Hoffman KL. The Psychopharmacology of Obsessive-Compulsive Disorder: A Preclinical Roadmap. Pharmacol Rev 2020; 72:80-151. [PMID: 31826934 DOI: 10.1124/pr.119.017772] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review evaluates current knowledge about obsessive-compulsive disorder (OCD), with the goal of providing a roadmap for future directions in research on the psychopharmacology of the disorder. It first addresses issues in the description and diagnosis of OCD, including the structure, measurement, and appropriate description of the disorder and issues of differential diagnosis. Current pharmacotherapies for OCD are then reviewed, including monotherapy with serotonin reuptake inhibitors and augmentation with antipsychotic medication and with psychologic treatment. Neuromodulatory therapies for OCD are also described, including psychosurgery, deep brain stimulation, and noninvasive brain stimulation. Psychotherapies for OCD are then reviewed, focusing on behavior therapy, including exposure and response prevention and cognitive therapy, and the efficacy of these interventions is discussed, touching on issues such as the timing of sessions, the adjunctive role of pharmacotherapy, and the underlying mechanisms. Next, current research on the neurobiology of OCD is examined, including work probing the role of various neurotransmitters and other endogenous processes and etiology as clues to the neurobiological fault that may underlie OCD. A new perspective on preclinical research is advanced, using the Research Domain Criteria to propose an adaptationist viewpoint that regards OCD as the dysfunction of a normal motivational system. A systems-design approach introduces the security motivation system (SMS) theory of OCD as a framework for research. Finally, a new perspective on psychopharmacological research for OCD is advanced, exploring three approaches: boosting infrastructure facilities of the brain, facilitating psychotherapeutic relearning, and targeting specific pathways of the SMS network to fix deficient SMS shut-down processes. SIGNIFICANCE STATEMENT: A significant proportion of patients with obsessive-compulsive disorder (OCD) do not achieve remission with current treatments, indicating the need for innovations in psychopharmacology for the disorder. OCD may be conceptualized as the dysfunction of a normal, special motivation system that evolved to manage the prospect of potential danger. This perspective, together with a wide-ranging review of the literature, suggests novel directions for psychopharmacological research, including boosting support systems of the brain, facilitating relearning that occurs in psychotherapy, and targeting specific pathways in the brain that provide deficient stopping processes in OCD.
Collapse
Affiliation(s)
- Henry Szechtman
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Brian H Harvey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Erik Z Woody
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| | - Kurt Leroy Hoffman
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada (H.S.); SAMRC Unit on Risk Resilience in Mental Disorders, Department of Psychiatry, University of Cape Town, and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University (Potchefstroom Campus), Potchefstroom, South Africa (B.H.H.); Department of Psychology, University of Waterloo, Waterloo, Ontario, Canada (E.Z.W.); and Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico (K.L.H.)
| |
Collapse
|
48
|
Tremblay ME, Cookson MR, Civiero L. Glial phagocytic clearance in Parkinson's disease. Mol Neurodegener 2019; 14:16. [PMID: 30953527 PMCID: PMC6451240 DOI: 10.1186/s13024-019-0314-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/15/2019] [Indexed: 12/21/2022] Open
Abstract
An emerging picture suggests that glial cells' loss of beneficial roles or gain of toxic functions can contribute to neurodegenerative conditions. Among glial cells, microglia and astrocytes have been shown to play phagocytic roles by engulfing synapses, apoptotic cells, cell debris, and released toxic proteins. As pathogenic protein accumulation is a key feature in Parkinson's disease (PD), compromised phagocytic clearance might participate in PD pathogenesis. In contrast, enhanced, uncontrolled and potentially toxic glial clearance capacity could contribute to synaptic degeneration. Here, we summarize the current knowledge of the molecular mechanisms underlying microglial and astrocytic phagocytosis, focusing on the possible implication of phagocytic dysfunction in neuronal degeneration. Several endo-lysosomal proteins displaying genetic variants in PD are highly expressed by microglia and astrocytes. We also present the evidence that lysosomal defects can affect phagocytic clearance and discuss the therapeutic relevance of restoring or enhancing lysosomal function in PD.
Collapse
Affiliation(s)
- Marie-Eve Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec, QC Canada
- Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, QC Canada
| | - Mark R. Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD USA
| | - Laura Civiero
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
| |
Collapse
|
49
|
Nasrolahi A, Safari F, Farhoudi M, Khosravi A, Farajdokht F, Bastaminejad S, Sandoghchian Shotorbani S, Mahmoudi J. Immune system and new avenues in Parkinson’s disease research and treatment. Rev Neurosci 2019; 30:709-727. [DOI: 10.1515/revneuro-2018-0105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022]
Abstract
Abstract
Parkinson’s disease (PD) is a progressive neurological disorder characterized by degeneration of dopaminergic neurons in the substantia nigra. However, although 200 years have now passed since the primary clinical description of PD by James Parkinson, the etiology and mechanisms of neuronal loss in this disease are still not fully understood. In addition to genetic and environmental factors, activation of immunologic responses seems to have a crucial role in PD pathology. Intraneuronal accumulation of α-synuclein (α-Syn), as the main pathological hallmark of PD, potentially mediates initiation of the autoimmune and inflammatory events through, possibly, auto-reactive T cells. While current therapeutic regimens are mainly used to symptomatically suppress PD signs, application of the disease-modifying therapies including immunomodulatory strategies may slow down the progressive neurodegeneration process of PD. The aim of this review is to summarize knowledge regarding previous studies on the relationships between autoimmune reactions and PD pathology as well as to discuss current opportunities for immunomodulatory therapy.
Collapse
Affiliation(s)
- Ava Nasrolahi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz 51666-14756 , Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Fatemeh Safari
- Departmant of Medical Biotechnology, School of Advanced Medical Sciences and Technologies , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz 51666-14756 , Iran
| | - Afra Khosravi
- Department of Immunology, Faculty of Medicine , Ilam University of Medical Sciences , Ilam , Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz 51666-14756 , Iran
| | - Saiyad Bastaminejad
- Department of Biochemistry and Molecular Medicine, School of Medicine , Ilam University of Medical Sciences , Ilam , Iran
| | | | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , P.O. 51666-14756, Tabriz , Iran , e-mail:
| |
Collapse
|
50
|
Nolan RA, Muir R, Runner K, Haddad EK, Gaskill PJ. Role of Macrophage Dopamine Receptors in Mediating Cytokine Production: Implications for Neuroinflammation in the Context of HIV-Associated Neurocognitive Disorders. J Neuroimmune Pharmacol 2018; 14:134-156. [PMID: 30519866 DOI: 10.1007/s11481-018-9825-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022]
Abstract
Despite the success of combination anti-retroviral therapy (cART), around 50% of HIV-infected individuals still display a variety of neuropathological and neurocognitive sequelae known as NeuroHIV. Current research suggests these effects are mediated by long-term changes in CNS function in response to chronic infection and inflammation, and not solely due to active viral replication. In the post-cART era, drug abuse is a major risk-factor for the development of NeuroHIV, and increases extracellular dopamine in the CNS. Our lab has previously shown that dopamine can increase HIV infection of primary human macrophages and increase the production of inflammatory cytokines, suggesting that elevated dopamine could enhance the development of HIV-associated neuropathology. However, the precise mechanism(s) by which elevated dopamine could exacerbate NeuroHIV, particularly in chronically-infected, virally suppressed individuals remain unclear. To determine the connection between dopaminergic alterations and HIV-associated neuroinflammation, we have examined the impact of dopamine exposure on macrophages from healthy and virally suppressed, chronically infected HIV patients. Our data show that dopamine treatment of human macrophages isolated from healthy and cART-treated donors promotes production of inflammatory mediators including IL-1β, IL-6, IL-18, CCL2, CXCL8, CXCL9, and CXCL10. Furthermore, in healthy individuals, dopamine-mediated modulation of specific cytokines is correlated with macrophage expression of dopamine-receptor transcripts, particularly DRD5, the most highly-expressed dopamine-receptor subtype. Overall, these data will provide more understanding of the role of dopamine in the development of NeuroHIV, and may suggest new molecules or pathways that can be useful as therapeutic targets during HIV infection.
Collapse
Affiliation(s)
- R A Nolan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - R Muir
- Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - K Runner
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - E K Haddad
- Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|