1
|
Xue F, Zhang M, Zhao RY, Wang XW, Gu Y, Yang Y, Chen WF. Dectin-1 participates in neuroinflammation and dopaminergic neurodegeneration through synergistic signaling crosstalk with TLR4. Brain Behav Immun 2025; 126:260-273. [PMID: 39978698 DOI: 10.1016/j.bbi.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Neuroinflammation mediated by microglial activation plays a prominent role in the pathogenesis of Parkinson's disease (PD). Dendritic cell-associated C-type lectin-1 (Dectin-1) is a pattern recognition receptor that is involved in innate immunity. However, the role of Dectin-1 on dopaminergic neuronal damage remains unclear. Our results demonstrated that the expression of Dectin-1 was significantly increased in the microglia of the LPS-induced PD mouse model. Inhibition of Dectin-1 by laminarin (LAM) attenuated LPS-induced dopaminergic neuronal damage in substantia nigra (SN) and behavioral deficits and promoted the phenotypic transformation of microglia from M1 to M2. Moreover, inhibition or knockdown of Dectin-1 significantly decreased LPS-induced phosphorylation of Syk and P65 as well as the production of COX-2 and iNOS in BV2 cells. Knockdown of Syk also significantly decreased LPS-induced protein expressions of COX-2 and iNOS. Mechanistically, both TLR4 inhibitor and NF-κB inhibitor could antagonize LPS-induced Dectin-1 expression. Chromatin immunoprecipitation (ChIP) assays showed a physical binding of NF-κB/P65 to Dectin-1 promoter, which further indicated the regulatory effect of toll-like receptor 4 (TLR4)/NF-κB signaling pathway on Dectin-1 expression. Furthermore, the present study provided the first evidence that Dectin-1 activation by hot-alkali treated depleted zymosan (d-Zymosan) could induce dopaminergic neurotoxicity and motor dysfunction, and promote up-regulation of TLR4, iNOS and Iba-1 in C57BL/6J mice. In conclusion, Dectin-1-Syk synergistic signaling crosstalk with TLR4/NF-κB promotes and maintains inflammatory phenotypes of M1 microglia which induces dopaminergic neuronal damage in SN. These findings provide novel insights into the pivotal role of Dectin-1 in neuroinflammation, suggesting its potential as a novel therapeutic target for PD.
Collapse
Affiliation(s)
- Feng Xue
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China; Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Mei Zhang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Rui-Yue Zhao
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xiao-Wen Wang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Yu Gu
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Ye Yang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, China
| | - Wen-Fang Chen
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
2
|
Tieu K, Salehe SS, Brown HJ. Toxin-Induced Animal Models of Parkinson's Disease. Cold Spring Harb Perspect Med 2025; 15:a041643. [PMID: 38951030 PMCID: PMC11875089 DOI: 10.1101/cshperspect.a041643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
The debilitating motor symptoms of Parkinson's disease (PD) result primarily from the degenerative nigrostriatal dopaminergic pathway. To elucidate pathogenic mechanisms and evaluate therapeutic strategies for PD, numerous animal models have been developed. Understanding the strengths and limitations of these models can significantly impact the choice of model, experimental design, and data interpretation. Herein, we systematically review the literature over the past decade. Some models no longer serve the purpose of PD models. The primary objectives of this review are: First, to assist new investigators in navigating through available animal models and making appropriate selections based on the objective of the study. Emphasis will be placed on common toxin-induced murine models. And second, to provide an overview of basic technical requirements for assessing the nigrostriatal pathway's pathology, structure, and function.
Collapse
Affiliation(s)
- Kim Tieu
- Department of Environmental Health Sciences, Florida International University, Miami, Florida 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, USA
| | - Said S Salehe
- Department of Environmental Health Sciences, Florida International University, Miami, Florida 33199, USA
| | - Harry J Brown
- Department of Environmental Health Sciences, Florida International University, Miami, Florida 33199, USA
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, USA
| |
Collapse
|
3
|
Breznik L, Daurer M, Rabl R, Loeffler T, Etxeberria-Rekalde E, Neddens J, Flunkert S, Prokesch M. Motor deficits and brain pathology in the Parkinson's disease mouse model hA53Ttg. Front Neurosci 2024; 18:1462041. [PMID: 39371610 PMCID: PMC11450652 DOI: 10.3389/fnins.2024.1462041] [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: 07/09/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
Background Parkinson's disease (PD) is a debilitating neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons and the accumulation of α-synuclein (α-syn) aggregates. The A53T missense point mutation occurs in autosomal dominant familial PD and has been found to promote the aggregation of α-syn. To investigate the role of the A53T mutation in PD, researchers have developed various mouse models with this mutation. Objective We therefore conducted a comprehensive characterization of the tg(THY1-SNCA*A53T)M53Sud mouse model (hA53Ttg mice) for its motor and pathological features. Methods hA53Ttg mice were tested for motor impairments in a series of motor tests at 2, 4 or 6 months of age. Human α-syn and α-syn pSer129, as well as GFAP and Iba1 signal were labeled and quantified in the cortex, hippocampus, and brainstem. Neurofilament light chain (NF-L) levels were measured in the cerebrospinal fluid (CSF) and plasma. Ex vivo analyses were performed at the age of 2, 4, 6, and 10 months. Results Behavioral tests revealed early muscle weakness and motor impairments that progressed with age. Immunohistochemical analyses demonstrated elevated levels of human α-syn and α-syn pSer129 in all evaluated brain regions. α-syn pSer129 labeling further revealed fiber-like structures in the cortex of older animals. Neuroinflammation was observed in an age-dependent manner. Biochemical evaluation revealed elevated NF-L levels in the plasma and CSF. Overall, our findings highlight the value of hA53Ttg mice in modeling PD-associated pathologies that closely resemble those observed in PD patients. Conclusion Our results thus suggest that hA53Ttg mice are a useful tool for studying the underlying mechanisms of PD.
Collapse
|
4
|
Laursen ALS, Olesen MV, Folke J, Brudek T, Knecht LH, Sotty F, Lambertsen KL, Fog K, Dalgaard LT, Aznar S. Systemic inflammation activates coagulation and immune cell infiltration pathways in brains with propagating α-synuclein fibril aggregates. Mol Cell Neurosci 2024; 129:103931. [PMID: 38508542 DOI: 10.1016/j.mcn.2024.103931] [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: 01/04/2024] [Revised: 02/15/2024] [Accepted: 03/13/2024] [Indexed: 03/22/2024] Open
Abstract
Synucleinopathies are a group of diseases characterized by brain aggregates of α-synuclein (α-syn). The gradual accumulation of α-syn and the role of inflammation in early-stage pathogenesis remain poorly understood. We explored this interaction by inducing chronic inflammation in a common pre-clinical synucleinopathy mouse model. Three weeks post unilateral intra-striatal injections of human α-syn pre-formed fibrils (PFF), mice underwent repeated intraperitoneal injections of 1 mg/ml lipopolysaccharide (LPS) for 3 weeks. Histological examinations of the ipsilateral site showed phospho-α-syn regional spread and LPS-induced neutrophil recruitment to the brain vasculature. Biochemical assessment of the contralateral site confirmed spreading of α-syn aggregation to frontal cortex and a rise in intracerebral TNF-α, IL-1β, IL-10 and KC/GRO cytokines levels due to LPS. No LPS-induced exacerbation of α-syn pathology load was observed at this stage. Proteomic analysis was performed contralateral to the PFF injection site using LC-MS/MS. Subsequent downstream Reactome Gene-Set Analysis indicated that α-syn pathology alters mitochondrial metabolism and synaptic signaling. Chronic LPS-induced inflammation further lead to an overrepresentation of pathways related to fibrin clotting as well as integrin and B cell receptor signaling. Western blotting confirmed a PFF-induced increase in fibrinogen brain levels and a PFF + LPS increase in Iba1 levels, indicating activated microglia. Splenocyte profiling revealed changes in T and B cells, monocytes, and neutrophils populations due to LPS treatment in PFF injected animals. In summary, early α-syn pathology impacts energy homeostasis pathways, synaptic signaling and brain fibrinogen levels. Concurrent mild systemic inflammation may prime brain immune pathways in interaction with peripheral immunity.
Collapse
Affiliation(s)
- Anne-Line Strange Laursen
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark; Department of Science and Environment, Roskilde University, Universitetsvej 1, DK-4000, Roskilde, Denmark.
| | - Mikkel Vestergaard Olesen
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | - Jonas Folke
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | - Tomasz Brudek
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | - Luisa Harriet Knecht
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| | | | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsløwsvej 21-25, DK-5000, Odense, Denmark; Department of Neurology, Odense University Hospital, J.B. Winsløwsvej 4, Odense, Denmark; BRIDGE - Brain-Research-Inter-Disciplinary Guided Excellence, Department of Clinical Institute, University of Southern Denmark, Winsløwparken 19, Odense, Denmark.
| | - Karina Fog
- H. Lundbeck A/S, Ottiliavej 9, DK-2500, Valby, Denmark.
| | - Louise Torp Dalgaard
- Department of Science and Environment, Roskilde University, Universitetsvej 1, DK-4000, Roskilde, Denmark.
| | - Susana Aznar
- Centre for Neuroscience & Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 6B, DK-2400, Copenhagen, NV, Denmark; Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Nielsine Nielsens Vej 4B, DK-2400, Copenhagen, NV, Denmark.
| |
Collapse
|
5
|
Monteiro Neto JR, Lima VDA, Follmer C. Fibrillation of α-synuclein triggered by bacterial endotoxin and lipid vesicles is modulated by N-terminal acetylation and familial Parkinson's disease mutations. FEBS J 2024; 291:1151-1167. [PMID: 38069536 DOI: 10.1111/febs.17027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 11/03/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023]
Abstract
It has been hypothesized that --Parkinson's disease (PD) may be initiated in the gastrointestinal tract, before manifesting in the central nervous system. In this respect, it was demonstrated that lipopolysaccharide (LPS), an endotoxin from gram-negative bacteria, accelerates the in vitro formation of α-synuclein (aSyn) fibrils, whose intracellular deposits is a histological hallmark of the degeneration of dopaminergic neurons in PD. Herein, N-terminal acetylation and missense mutations of aSyn (A30P, A53T, E46K, H50Q and G51D) linked to rare, early-onset forms of familial PD were investigated regarding their effect on aSyn aggregation stimulated by either LPS or small unilamellar lipid vesicles (SUVs). Our findings indicated that LPS as well as SUVs induce the fibrillation of N-terminally acetylated wild-type aSyn (Ac-aSyn-WT) more remarkably than the non-acetylated protein, while the LPS-free protein alone did not undergo fibrillation under our assay conditions. In addition, with the exception of A30P, PD mutations increased the fibrillation of Ac-aSyn in the presence of LPS compared with Ac-aSyn-WT. The most pronounced effect of LPS was noticed for A53T, as observed when either Thioflavin-T or JC-1 were used as fluorescent probes for fibrils. Overall, our results suggest for the first time the existence of a synergy between LPS and PD mutations/N-terminal acetylation toward aSyn fibrillation.
Collapse
Affiliation(s)
- José Raphael Monteiro Neto
- Laboratory of Biological Chemistry of Neurodegenerative Disorders, Institute of Chemistry, Federal University of Rio de Janeiro, Brazil
| | - Vanderlei de Araújo Lima
- Laboratory of Biological Chemistry of Neurodegenerative Disorders, Institute of Chemistry, Federal University of Rio de Janeiro, Brazil
| | - Cristian Follmer
- Laboratory of Biological Chemistry of Neurodegenerative Disorders, Institute of Chemistry, Federal University of Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Sanfeliu C, Bartra C, Suñol C, Rodríguez-Farré E. New insights in animal models of neurotoxicity-induced neurodegeneration. Front Neurosci 2024; 17:1248727. [PMID: 38260026 PMCID: PMC10800989 DOI: 10.3389/fnins.2023.1248727] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
The high prevalence of neurodegenerative diseases is an unintended consequence of the high longevity of the population, together with the lack of effective preventive and therapeutic options. There is great pressure on preclinical research, and both old and new models of neurodegenerative diseases are required to increase the pipeline of new drugs for clinical testing. We review here the main models of neurotoxicity-based animal models leading to central neurodegeneration. Our main focus was on studying how changes in neurotransmission and neuroinflammation, mainly in rodent models, contribute to harmful processes linked to neurodegeneration. The majority of the models currently in use mimic Parkinson's disease (PD) and Alzheimer's disease (AD), which are the most common neurodegenerative conditions in older adults. AD is the most common age-related dementia, whereas PD is the most common movement disorder with also cases of dementia. Several natural toxins and xenobiotic agents induce dopaminergic neurodegeneration and can reproduce neuropathological traits of PD. The literature analysis of MPTP, 6-OH-dopamine, and rotenone models suggested the latter as a useful model when specific doses of rotenone were administrated systemically to C57BL/6 mice. Cholinergic neurodegeneration is mainly modelled with the toxin scopolamine, which is a useful rodent model for the screening of protective drugs against cognitive decline and AD. Several agents have been used to model neuroinflammation-based neurodegeneration and dementia in AD, including lipopolysaccharide (LPS), streptozotocin, and monomeric C-reactive protein. The bacterial agent LPS makes a useful rodent model for testing anti-inflammatory therapies to halt the development and severity of AD. However, neurotoxin models might be more useful than genetic models for drug discovery in PD but that is not the case in AD where they cannot beat the new developments in transgenic mouse models. Overall, we should work using all available models, either in vivo, in vitro, or in silico, considering the seriousness of the moment and urgency of developing effective drugs.
Collapse
Affiliation(s)
- Coral Sanfeliu
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Clara Bartra
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- PhD Program in Biotechnology, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Cristina Suñol
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Eduard Rodríguez-Farré
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
7
|
Long H, Zhu W, Wei L, Zhao J. Iron homeostasis imbalance and ferroptosis in brain diseases. MedComm (Beijing) 2023; 4:e298. [PMID: 37377861 PMCID: PMC10292684 DOI: 10.1002/mco2.298] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 06/29/2023] Open
Abstract
Brain iron homeostasis is maintained through the normal function of blood-brain barrier and iron regulation at the systemic and cellular levels, which is fundamental to normal brain function. Excess iron can catalyze the generation of free radicals through Fenton reactions due to its dual redox state, thus causing oxidative stress. Numerous evidence has indicated brain diseases, especially stroke and neurodegenerative diseases, are closely related to the mechanism of iron homeostasis imbalance in the brain. For one thing, brain diseases promote brain iron accumulation. For another, iron accumulation amplifies damage to the nervous system and exacerbates patients' outcomes. In addition, iron accumulation triggers ferroptosis, a newly discovered iron-dependent type of programmed cell death, which is closely related to neurodegeneration and has received wide attention in recent years. In this context, we outline the mechanism of a normal brain iron metabolism and focus on the current mechanism of the iron homeostasis imbalance in stroke, Alzheimer's disease, and Parkinson's disease. Meanwhile, we also discuss the mechanism of ferroptosis and simultaneously enumerate the newly discovered drugs for iron chelators and ferroptosis inhibitors.
Collapse
Affiliation(s)
- Haining Long
- Department of Diagnostic and Interventional RadiologyShanghai Sixth People’s Hospital Afliated to Shanghai Jiao Tong University School
of MedicineShanghaiChina
| | - Wangshu Zhu
- Department of Diagnostic and Interventional RadiologyShanghai Sixth People’s Hospital Afliated to Shanghai Jiao Tong University School
of MedicineShanghaiChina
| | - Liming Wei
- Department of Diagnostic and Interventional RadiologyShanghai Sixth People’s Hospital Afliated to Shanghai Jiao Tong University School
of MedicineShanghaiChina
| | - Jungong Zhao
- Department of Diagnostic and Interventional RadiologyShanghai Sixth People’s Hospital Afliated to Shanghai Jiao Tong University School
of MedicineShanghaiChina
| |
Collapse
|
8
|
Oliynyk Z, Rudyk M, Dovbynchuk T, Dzubenko N, Tolstanova G, Skivka L. Inflammatory hallmarks in 6-OHDA- and LPS-induced Parkinson's disease in rats. Brain Behav Immun Health 2023; 30:100616. [PMID: 37096171 PMCID: PMC10121378 DOI: 10.1016/j.bbih.2023.100616] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/08/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, affecting more than 1% of aged people. PD, which was previously identified as movement disorder, now is recognized as a multi-factorial systemic disease with important pathogenetic and pathophysiological role of inflammation. Reproducing local and systemic inflammation, which is inherent in PD, in animal models is essential for maximizing the translation of their potential to the clinic, as well as for developing putative anti-inflammatory neuroprotective agents. This study was aimed to compare activation patterns of microglia/macrophage population and systemic inflammation indices in rats with 6-Hydroxydopamine (6-OHDA)- and Lipopolysaccharide (LPS)-induced PD. Metabolic and phenotypic characteristics of microglia/macrophage population were examined by flow cytometry, systemic inflammatory markers were calculated using hematological parameters in 6-OHDA- and LPS-lesioned Wistar rats 29 days after the surgery. Microglia/macrophages from rats in both models exhibited pro-inflammatory metabolic shift. Nevertheless, in LPS-lesioned animals, highly increased proportion of CD80/86+ cells in microglia/macrophage population was registered alongside increased values of systemic inflammatory indices: neutrophil to lymphocyte ratio (NLR), derived neutrophil to lymphocyte ratio (dNLR), platelet to lymphocyte ratio and systemic immune inflammation index (SII). There was significant positive correlation between the count of CD80/86+ cells and systemic inflammatory indices in these animals. Microglia/macrophages from 6-OHDA-lesioned rats were characterized by the increased fraction of CD206+ cells alongside decreased proportion of CD80/86+ cells. No signs of systemic inflammation were observed. Negative correlation between quantitation characteristics of CD80/86+ cells and values of systemic inflammatory indices was registered. Collectively, our data show that LPS-PD model unlike 6-OHDA-PD replicates crosstalk between local and systemic inflammatory responses, which is inherent in PD pathogenesis and pathophysiology.
Collapse
Affiliation(s)
- Zhanna Oliynyk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv, 03022, Ukraine
| | - Mariia Rudyk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv, 03022, Ukraine
- Corresponding author. Microbiology and Immunology Department, ESC “Institute of Biology and Medicine”, Taras Shevchenko Kyiv National University, Kyiv, 2, Hlushkov Avenue, Kyiv, 03022, Ukraine.
| | - Taisa Dovbynchuk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv, 03022, Ukraine
| | - Nataliia Dzubenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv, 03022, Ukraine
| | - Ganna Tolstanova
- Educational and Scientific Institute of High Technologies, Taras Shevchenko University of Kyiv, 4g, Hlushkova Avenue, Kyiv, 03022, Ukraine
| | - Larysa Skivka
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv, 03022, Ukraine
| |
Collapse
|
9
|
Forloni G. Alpha Synuclein: Neurodegeneration and Inflammation. Int J Mol Sci 2023; 24:ijms24065914. [PMID: 36982988 PMCID: PMC10059798 DOI: 10.3390/ijms24065914] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Alpha-Synuclein (α-Syn) is one of the most important molecules involved in the pathogenesis of Parkinson's disease and related disorders, synucleinopathies, but also in several other neurodegenerative disorders with a more elusive role. This review analyzes the activities of α-Syn, in different conformational states, monomeric, oligomeric and fibrils, in relation to neuronal dysfunction. The neuronal damage induced by α-Syn in various conformers will be analyzed in relation to its capacity to spread the intracellular aggregation seeds with a prion-like mechanism. In view of the prominent role of inflammation in virtually all neurodegenerative disorders, the activity of α-Syn will also be illustrated considering its influence on glial reactivity. We and others have described the interaction between general inflammation and cerebral dysfunctional activity of α-Syn. Differences in microglia and astrocyte activation have also been observed when in vivo the presence of α-Syn oligomers has been combined with a lasting peripheral inflammatory effect. The reactivity of microglia was amplified, while astrocytes were damaged by the double stimulus, opening new perspectives for the control of inflammation in synucleinopathies. Starting from our studies in experimental models, we extended the perspective to find useful pointers to orient future research and potential therapeutic strategies in neurodegenerative disorders.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy
| |
Collapse
|
10
|
Huang B, Hu G, Zong X, Yang S, He D, Gao X, Liu D. α-Cyperone protects dopaminergic neurons and inhibits neuroinflammation in LPS-induced Parkinson's disease rat model via activating Nrf2/HO-1 and suppressing NF-κB signaling pathway. Int Immunopharmacol 2023; 115:109698. [PMID: 36634417 DOI: 10.1016/j.intimp.2023.109698] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/21/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Our previous study showed that α-Cyperone inhibited the inflammatory response triggered by activated microglia and protected dopaminergic neuron in in vitro cell model of Parkinson's disease (PD). It is unclear the effect of α-Cyperone in animal models of PD. In this study, our results indicated that α-Cyperone ameliorated motor dysfunction, protected dopaminergic neurons, and inhibited the reduction of dopamine and its metabolites in lipopolysaccharide (LPS)-induced PD rat model. Moreover, α-Cyperone suppressed the activation of microglia and the expression of neuroinflammatory factor (TNF-α, IL-6, IL-1β, iNOS, COX-2 and ROS). Furthermore, the molecular mechanism research revealed that α-Cyperone inhibited neuroinflammation and oxidative stress to exert protective effect in microglia by activating Nrf2/HO-1 and suppressing NF-κB signaling pathway. Moreover, α-Cyperone upregulated the expression of antioxidant enzymes (GCLC, GCLM and NQO1) in microglia. In conclusion, our study demonstrates α-Cyperone alleviates dopaminergic neurodegeneration by inhibiting neuroinflammation and oxidative stress in LPS-induced PD rat model via activating Nrf2/HO-1 and suppressing NF-κB signaling pathway.
Collapse
Affiliation(s)
- Bingxu Huang
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Guiqiu Hu
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaofeng Zong
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shuo Yang
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Dewei He
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Xiyu Gao
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Dianfeng Liu
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China.
| |
Collapse
|