1
|
Xie Y, Wang XY, Liu S, He ZY, Zhang H, Yu ZY, Xie MJ, Wang W. Transforming Growth Factor β1 Protects Against Ischemic Demyelination via Regulating Microglial Lipid Metabolism Pathway. Stroke 2025. [PMID: 40160039 DOI: 10.1161/strokeaha.124.048206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 01/13/2025] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Chronic cerebral hypoperfusion-induced white matter lesions are an important cause of vascular cognitive impairment in aging life. TGF-β1 (transforming growth factor β1) is widely recognized as a multifunctional cytokine participating in numerous pathophysiological processes in the central nervous system. In this study, we aimed to evaluate the neuroprotective potentials of TGF-β1 in ischemic white matter lesions. METHODS A mouse model of bilateral common carotid artery stenosis was established to imitate the ischemic white matter lesions. The agonist of the TGF-β1 pathway was continuously applied via intraperitoneal injection. The Morris water maze test and gait analysis system were used to assess the cognitive and gait disorders in modeling mice. The Luxol fast blue staining, immunofluorescence, and electron microscopy were conducted to determine the severity of demyelinating lesions, microglial activation, and dysfunction of the autophagy-lysosomal pathway in microglia. Furthermore, primary cultured microglia were exposed to extracted myelin debris and TGF-β1 in vitro to explore the underlying mechanisms. RESULTS As evaluated by behavioral tests, TGF-β1 significantly alleviated the cognitive dysfunction and gait disorder in bilateral common carotid artery stenosis-modeling mice. The demyelinating lesion and remyelination process were also found to be highly improved by activation of the TGF-β1 pathway. The results of immunostaining and electron microscopy showed that TGF-β1 could ameliorate microglial activation and the dysfunction of lipid metabolism in myelin-engulfed microglia. Mechanistically, in primary cultured microglia exposed to myelin debris, administration of TGF-β1 notably mitigated the inflammatory response and accumulation of intracellular lipid droplets via promoting the lipid droplets degradation in the autophagy-lysosomal pathway, as quantified by flow cytometry, immunostaining, Western blot, etc. Yet, the application of autophagy inhibitor (3-methyladenine) significantly reversed the above anti-inflammatory effects of TGF-β1. CONCLUSIONS TGF-β1 relieved cognitive deficit, demyelinating lesions, and microglia-mediated neuroinflammation in bilateral common carotid artery stenosis modeling by reducing abnormal lipid droplet accumulation and dysfunction of the autophagy-lysosomal pathway in microglia. Clinically, staged activation of the TGF-β1 pathway may become a potential target and promising treatment for ischemic white matter lesions and vascular cognitive impairment.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
| | - Xin-Yue Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
| | - Shuai Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China. (S.L.)
| | - Zi-Yu He
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
| | - Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
| | - Zhi-Yuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
| | - Min-Jie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. (W.W.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.-Y.W., Z.-Y.H., H.Z., Z.-Y.Y., M.-J.X., W.W.)
| |
Collapse
|
2
|
Mincheva G, Felipo V, Moreno-Manzano V, Benítez-Páez A, Llansola M. Extracellular vesicles from mesenchymal stem cells alter gut microbiota and improve neuroinflammation and motor impairment in rats with mild liver damage. Neurotherapeutics 2024; 21:e00445. [PMID: 39242290 PMCID: PMC11585882 DOI: 10.1016/j.neurot.2024.e00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
Gut microbiota perturbation and motor dysfunction have been reported in steatosis patients. Rats with mild liver damage (MLD) show motor dysfunction mediated by neuroinflammation and altered GABAergic neurotransmission in the cerebellum. The extracellular vesicles (EV) from mesenchymal stem cells (MSC) have emerged as a promising therapeutic proxy whose molecular basis relies partly upon TGFβ action. This study aimed to assess if MSC-EVs improve motor dysfunction in rats with mild liver damage and analyze underlying mechanisms, including the role of TGFβ, cerebellar neuroinflammation and gut microbiota. MLD in rats was induced by carbon tetrachloride administration and EVs from normal (C-EVs) or TGFβ-siRNA treated MSCs (T-EV) were injected. Motor coordination, locomotor gait, neuroinflammation and TNF-α-activated pathways modulating GABAergic neurotransmission in the cerebellum, microbiota composition in feces and microbial-derived metabolites in plasma were analyzed. C-EVs reduced glial and TNFα-P2X4-BDNF-TrkB pathway activation restoring GABAergic neurotransmission in the cerebellum and improving motor coordination and all the altered gait parameters. T-EVs also improved motor coordination and some gait parameters, but the mechanisms involved differed from those of C-EVs. MLD rats showed increased content of some Bacteroides species in feces, correlating with decreased kynurenine aside from motor alterations. These alterations were all normalized by C-EVs, whereas T-EVs only restored kynurenine levels. Our results support the value of MSC-EVs on improving motor dysfunction in MLD and unveil a possible mechanism by which altered microbiota may contribute to neuroinflammation and motor impairment. Some of the underlying mechanisms are TGFβ-dependent.
Collapse
Affiliation(s)
- Gergana Mincheva
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain
| | - Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Laboratory, Centro Investigación Príncipe Felipe, Valencia, Spain
| | - Alfonso Benítez-Páez
- Host-Microbe Interactions in Metabolic Health Laboratory, Centro de Investigación Principe Felipe, Valencia, Spain; Microbiome, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology (IATA-CSIC). Paterna-Valencia, Spain..
| | - Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
3
|
Sun X, Gu R, Bai J. Differentiation and regulation of CD4 + T cell subsets in Parkinson's disease. Cell Mol Life Sci 2024; 81:352. [PMID: 39153043 PMCID: PMC11335276 DOI: 10.1007/s00018-024-05402-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, and its hallmark pathological features are the loss of dopaminergic (DA) neurons in the midbrain substantia nigra pars compacta (SNpc) and the accumulation of alpha-synuclein (α-syn). It has been shown that the integrity of the blood-brain barrier (BBB) is damaged in PD patients, and a large number of infiltrating T cells and inflammatory cytokines have been detected in the cerebrospinal fluid (CSF) and brain parenchyma of PD patients and PD animal models, including significant change in the number and proportion of different CD4+ T cell subsets. This suggests that the neuroinflammatory response caused by CD4+ T cells is an important risk factor for the development of PD. Here, we systematically review the differentiation of CD4+ T cell subsets, and focus on describing the functions and mechanisms of different CD4+ T cell subsets and their secreted cytokines in PD. We also summarize the current immunotherapy targeting CD4+ T cells with a view to providing assistance in the diagnosis and treatment of PD.
Collapse
Affiliation(s)
- Xiaowei Sun
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
- Southwest United Graduate School, Kunming, 650500, China
| | - Rou Gu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
- Southwest United Graduate School, Kunming, 650500, China.
| |
Collapse
|
4
|
Lepiarz-Raba I, Hidayat T, Hannan AJ, Jawaid A. Potential Alzheimer's disease drug targets identified through microglial biology research. Expert Opin Drug Discov 2024; 19:587-602. [PMID: 38590098 DOI: 10.1080/17460441.2024.2335210] [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: 01/20/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024]
Abstract
INTRODUCTION Microglia, the primary immune cells in the brain, play multifaceted roles in Alzheimer's disease (AD). Microglia can potentially mitigate the pathological progression of AD by clearing amyloid beta (Aβ) deposits in the brain and through neurotrophic support. In contrast, disproportionate activation of microglial pro-inflammatory pathways, as well as excessive elimination of healthy synapses, can exacerbate neurodegeneration in AD. The challenge, therefore, lies in discerning the precise regulation of the contrasting microglial properties to harness their therapeutic potential in AD. AREAS COVERED This review examines the evidence relevant to the disease-modifying effects of microglial manipulators in AD preclinical models. The deleterious pro-inflammatory effects of microglia in AD can be ameliorated via direct suppression or indirectly through metabolic manipulation, epigenetic targeting, and modulation of the gut-brain axis. Furthermore, microglial clearance of Aβ deposits in AD can be enhanced via strategically targeting microglial membrane receptors, lysosomal functions, and metabolism. EXPERT OPINION Given the intricate and diverse nature of microglial responses throughout the course of AD, therapeutic interventions directed at microglia warrant a tactical approach. This could entail employing therapeutic regimens, which concomitantly suppress pro-inflammatory microglial responses while selectively enhancing Aβ phagocytosis.
Collapse
Affiliation(s)
- Izabela Lepiarz-Raba
- Laboratory for Translational Research in Exposures and Neuropsychiatric Disorders (TREND), Braincity: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Taufik Hidayat
- Laboratory for Translational Research in Exposures and Neuropsychiatric Disorders (TREND), Braincity: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Ali Jawaid
- Laboratory for Translational Research in Exposures and Neuropsychiatric Disorders (TREND), Braincity: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
5
|
Pinjala P, Tryphena KP, Kulkarni A, Goswami PG, Khatri DK. Dimethyl Fumarate Exerts a Neuroprotective Effect by Enhancing Mitophagy via the NRF2/BNIP3/PINK1 Axis in the MPP + Iodide-Induced Parkinson's Disease Mice Model. J Alzheimers Dis Rep 2024; 8:329-344. [PMID: 38405353 PMCID: PMC10894611 DOI: 10.3233/adr-230128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/28/2023] [Indexed: 02/27/2024] Open
Abstract
Background Parkinson's disease (PD) is a progressive neurodegenerative disorder linked to the loss of dopaminergic neurons in the substantia nigra. Mitophagy, mitochondrial selective autophagy, is critical in maintaining mitochondrial and subsequently neuronal homeostasis. Its impairment is strongly implicated in PD and is associated with accelerated neurodegeneration. Objective To study the positive effect of dimethyl fumarate (DMF) on mitophagy via the NRF2/BNIP3/PINK1 axis activation in PD disease models. Methods The neuroprotective effect of DMF was explored in in vitro and in vivo PD models. MTT assay was performed to determine the DMF dose followed by JC-1 assay to study its mitoprotective effect in MPP+ exposed SHSY5Y cells. For the in vivo study, C57BL/6 mice were divided into six groups: Normal Control (NC), Disease Control (DC), Sham (Saline i.c.v.), Low Dose (MPP+ iodide+DMF 15 mg/kg), Mid Dose (MPP+ iodide+DMF 30 mg/kg), and High Dose (MPP+ iodide+DMF 60 mg/kg). The neuroprotective effect of DMF was assessed by performing rotarod, open field test, and pole test, and biochemical parameter analysis using immunofluorescence, western blot, and RT-PCR. Results DMF treatment significantly alleviated the loss of TH positive dopaminergic neurons and enhanced mitophagy by increasing PINK1, Parkin, BNIP3, and LC3 levels in the MPP+ iodide-induced PD mice model. DMF treatment groups showed good locomotor activity and rearing time when compared to the DC group. Conclusions DMF confers neuroprotection by activating the BNIP3/PINK1/Parkin pathway, enhancing the autophagosome formation via LC3, and improving mitophagy in PD models, and could be a potential therapeutic option in PD.
Collapse
Affiliation(s)
- Poojitha Pinjala
- Department of Pharmacology and Toxicology, Molecular and Cellular Neuroscience Lab, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Kamatham Pushpa Tryphena
- Department of Pharmacology and Toxicology, Molecular and Cellular Neuroscience Lab, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Amrita Kulkarni
- Department of Pharmacology and Toxicology, Molecular and Cellular Neuroscience Lab, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Prince Giri Goswami
- Department of Pharmacology and Toxicology, Molecular and Cellular Neuroscience Lab, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, Molecular and Cellular Neuroscience Lab, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
- Department of Pharmacology, Shobhaben Pratapbai Patel School of Pharmacy and Technology Management, SVKM’s Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be-University, Mumbai, India
| |
Collapse
|
6
|
Xie Y, Chen X, Wang X, Liu S, Chen S, Yu Z, Wang W. Transforming growth factor-β1 protects against white matter injury and reactive astrogliosis via the p38 MAPK pathway in rodent demyelinating model. J Neurochem 2024; 168:83-99. [PMID: 38183677 DOI: 10.1111/jnc.16037] [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: 08/22/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 01/08/2024]
Abstract
In central nervous system (CNS), demyelination is a pathological process featured with a loss of myelin sheaths around axons, which is responsible for the diseases of multiple sclerosis, neuromyelitis optica, and so on. Transforming growth factor-beta1 (TGF-β1) is a multifunctional cytokine participating in abundant physiological and pathological processes in CNS. However, the effects of TGF-β1 on CNS demyelinating disease and its underlying mechanisms are controversial and not well understood. Herein, we evaluated the protective potential of TGF-β1 in a rodent demyelinating model established by lysophosphatidylcholine (LPC) injection. It was identified that supplement of TGF-β1 evidently rescued the cognitive deficit and motor dysfunction in LPC modeling mice assessed by novel object recognition and balance beam behavioral tests. Besides, quantified by luxol fast blue staining, immunofluorescence, and western blot, administration of TGF-β1 was found to significantly ameliorate the demyelinating lesion and reactive astrogliosis by suppressing p38 MAPK pathway. Mechanistically, the results of in vitro experiments indicated that treatment of TGF-β1 could directly promote the differentiation and migration of cultured oligodendrocytes. Our study revealed that modulating TGF-β1 activity might serve as a promising and innovative therapeutic strategy in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Xuejiao Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyue Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Simiao Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Liu Y, Duan R, Li P, Zhang B, Liu Y. 3-N-butylphthalide attenuates neuroinflammation in rotenone-induced Parkinson's disease models via the cGAS-STING pathway. Int J Immunopathol Pharmacol 2024; 38:3946320241229041. [PMID: 38315064 PMCID: PMC10846052 DOI: 10.1177/03946320241229041] [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: 10/20/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Neuroinflammation is crucial in the onset and progression of dopaminergic neuron loss in Parkinson's disease (PD). We aimed to determine whether 3-N-Butylphthalide (NBP) can protect against PD by inhibiting the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway and the inflammatory response of microglia. MitoSOX/MitoTracker/Hoechst staining was used to detect the levels of mitochondrial reactive oxygen species (ROS) in BV2 cells. Quantitative Real-Time Polymerase Chain Reaction was used to measure the levels of free cytoplasmic mitochondrial DNA (mtDNA) in BV2 cells and mouse brain tissues. Behavioral impairments were assessed using rotarod, T-maze, and balance beam tests. Dopaminergic neurons and microglia were observed using immunohistochemical staining. Expression levels of cGAS, STING, nuclear factor kappa-B (NfκB), phospho- NfκB (p-NfκB), inhibitor of NfκBα (IκBα), and phospho-IκBα (p-IκBα) proteins in the substantia nigra and striatum were detected using Western Blot. NBP decreased mitochondrial ROS levels in rotenone-treated BV2 cells. NBP alleviated behavioral impairments and protected against rotenone-induced microgliosis and damage to dopaminergic neurons in the substantia nigra and striatum of rotenone-induced PD mice. NBP decreased rotenone-induced mtDNA leakage and mitigated neuroinflammation by inhibiting cGAS-STING pathway activation. NBP exhibited a protective effect in rotenone-induced PD models by significantly inhibiting the cGAS-STING pathway. Moreover, NBP can alleviate neuroinflammation, and is a potential therapeutic drug for alleviating clinical symptoms and delaying the progression of PD. This study provided insights for the potential role of NBP in PD therapy, potentially mitigating neurodegeneration, and consequently improving the quality of life and lifespan of patients with PD. The limitations are that we have not confirmed the exact mechanism by which NBP decreases mtDNA leakage, and this study was unable to observe the actual clinical therapeutic effect, so further cohort studies are required for validation.
Collapse
Affiliation(s)
- Yuqian Liu
- Qilu Hospital of Shandong University, Jinan, China
| | - Ruonan Duan
- Qilu Hospital of Shandong University, Jinan, China
| | - Peizheng Li
- Qilu Hospital of Shandong University, Jinan, China
| | - Bohan Zhang
- Qilu Hospital of Shandong University, Jinan, China
| | - Yiming Liu
- Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
8
|
Rubio-Osornio M, León CTGD, Montes S, Rubio C, Ríos C, Monroy A, Morales-Montor J. Repurposing Simvastatin in Parkinson's Disease Model: Protection Is throughout Modulation of the Neuro-Inflammatory Response in the Substantia nigra. Int J Mol Sci 2023; 24:10414. [PMID: 37445592 DOI: 10.3390/ijms241310414] [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: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 07/15/2023] Open
Abstract
Parkinson's disease is a neurodegenerative disorder characterized by oxidative stress and immune activation in the nigro-striatal pathway. Simvastatin regulates cholesterol metabolism and protects from atherosclerosis disease. Simvastatin-tween 80 was administered 7 days before sterotaxic intrastriatal administration of MPP+ (1-methyl-4-phenylpyridine) in rats. Fluorescent lipidic product formation, dopamine levels, and circling behavior were considered damage markers. Twenty-four hours and six days after, the animal group lesioned with MPP+ showed significant damage in relation to the control group. Animals pretreated with simvastatin significantly reduced the MPP+-induced damage compared to the MPP+ treated group. As apoptosis promotes neuroinflammation and neuronal degeneration in Parkinson's disease, and since there is not currently a proteomic map of the nigro-striatum of rats and assuming a high homology among the identified proteins in other rat tissues, we based the search for rat protein homologs related to the establishment of inflammation response. We demonstrate that most proteins related to inflammation decreased in the simvastatin-treated rats. Furthermore, differential expression of antioxidant enzymes in striated tissue of rat brains was found in response to simvastatin. These results suggest that simvastatin could prevent striatal MPP+-induced damage and, for the first time, suggest that the molecular mechanisms involved in this have a protective effect.
Collapse
Affiliation(s)
- Moisés Rubio-Osornio
- Departamento de Neuroquímica, Instituto Nacional de Neurología and Neurocirugía, Tlalpan, Ciudad de Mexico 14269, Mexico
| | - Carmen T Goméz-De León
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de Mexico 04510, Mexico
| | - Sergio Montes
- Unidad Académica Multidisciplinaria, Departamento de Farmacología, Universidad Autónoma de Tamaulipas, Reynosa Tamaulipas 88740, Mexico
| | - Carmen Rubio
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Tlalpan, Ciudad de Mexico 14269, Mexico
| | - Camilo Ríos
- Dirección de Investigación, Instituto Nacional de Rehabilitación, Tlalpan, Ciudad de Mexico 14389, Mexico
| | - Antonio Monroy
- Laboratorio de Neuroprotección, Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de Mexico 04510, Mexico
| |
Collapse
|
9
|
Scotton E, Casa PL, de Abreu FP, de Avila E Silva S, Wilges RLB, Rossetto MV, Géa LP, Rosa AR, Colombo R. Differentially regulated targets in the fast-acting antidepressant effect of (R)-ketamine: A systems biology approach. Pharmacol Biochem Behav 2023; 223:173523. [PMID: 36731751 DOI: 10.1016/j.pbb.2023.173523] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023]
Abstract
Approximately two-thirds of patients with major depressive disorder (MDD) fail to respond to conventional antidepressants, suggesting that additional mechanisms are involved in the MDD pathophysiology. In this scenario, the glutamatergic system represents a promising therapeutic target for treatment-resistant depression. To our knowledge, this is the first study using semantic approach with systems biology to identify potential targets involved in the fast-acting antidepressant effects of ketamine and its enantiomers as well as identifying specific targets of (R)-ketamine. We performed a systematic review, followed by a semantic analysis and functional gene enrichment to identify the main biological processes involved in the therapeutic effects of these agents. Protein-protein interaction networks were constructed, and the genes exclusively regulated by (R)-ketamine were explored. We found that the regulation of α-Amino-3-Hydroxy-5-Methyl-4-Isoxazolepropionic Acid (AMPA) receptor and N-methyl-d-aspartate (NMDA) receptor subunits-Postsynaptic Protein 95 (PSD-95), Brain Derived Neurotrophic Factor (BDNF), and Tyrosine Receptor Kinase B (TrkB) are shared by the three-antidepressant agents, reinforcing the central role of the glutamatergic system and neurogenesis on its therapeutic effects. Differential regulation of Transforming Growth Factor Beta 1 (TGF-β1) receptors-Mitogen-Activated Protein Kinases (MAPK's), Receptor Activator of Nuclear Factor-Kappa Beta Ligand (RANKL), and Serotonin Transporter (SERT) seems to be particularly involved in (R)-ketamine antidepressant effects. Our data helps further studies investigating the relationship between these targets and the mechanisms of (R)-ketamine and searching for other therapeutic compounds that share the regulation of these specific biomolecules. Ultimately, this study could contribute to improve the fast management of depressive-like symptoms with less detrimental side effects than ketamine and (S)-ketamine.
Collapse
Affiliation(s)
- Ellen Scotton
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Pharmacology Department and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Pedro Lenz Casa
- Institute of Biotechnology, Universidade de Caxias do Sul (UCS), Caxias do Sul, RS, Brazil.
| | | | | | - Renata Luiza Boff Wilges
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | | | - Luiza Paul Géa
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Adriane R Rosa
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Pharmacology Department and Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Rafael Colombo
- Institute of Biotechnology, Universidade de Caxias do Sul (UCS), Caxias do Sul, RS, Brazil.
| |
Collapse
|
10
|
Liu B, Wang Y, He D, Han G, Wang H, Lin Y, Zhang T, Yi C, Li H. LTBP1 Gene Expression in the Cerebral Cortex and its Neuroprotective Mechanism in Mice with Postischemic Stroke Epilepsy. Curr Pharm Biotechnol 2023; 24:317-329. [PMID: 35676846 DOI: 10.2174/1389201023666220608091511] [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: 02/08/2022] [Revised: 03/08/2022] [Accepted: 03/30/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study aimed at exploring the expression level of LTBP1 in the mouse model of epilepsy. The mechanism of LTBP1 in epileptic cerebral neural stem cells was deeply investigated to control the occurrence of epilepsy with neuroprotection. METHODS qRT-PCR was conducted for the expression levels of LTBP1 in clinical human epileptic tissues and neural stem cells, as well as normal cerebral tissues and neural stem cells. The mouse model of postischemic stroke epilepsy (PSE) was established by the middle cerebral artery occlusion (MCAO). Then, qRT-PCR was conducted again for the expression levels of LTBP1 in mouse epileptic tissues and neural stem cells as well as normal cerebral tissues and neural stem cells. The activation and inhibitory vectors of LTBP1 were constructed to detect the effects of LTBP1 on the proliferation of cerebral neural stem cells in the PSE model combined with CCK-8. Finally, Western blot was conducted for the specific mechanism of LTBP1 affecting the development of epileptic cells. RESULTS Racine score and epilepsy index of 15 mice showed epilepsy symptoms after the determination with MCAO, showing a successful establishment of the PSE model. LTBP1 expression in both diseased epileptic tissues and cells was higher than that in normal clinical epileptic tissues and cells. Meanwhile, qRT-PCR showed higher LTBP1 expression in both mouse epileptic tissues and their neural stem cells compared to that in normal tissues and cells. CCK-8 showed that the activation of LTBP1 stimulated the increased proliferative capacity of epileptic cells, while the inhibition of LTBP1 expression controlled the proliferation of epileptic cells. Western blot showed an elevated expression of TGFβ/SMAD signaling pathway-associated protein SMAD1/5/8 after activating LTBP1. The expression of molecular MMP-13 associated with the occurrence of inflammation was also activated. CONCLUSION LTBP1 can affect the changes in inflammation-related pathways by activating the TGFβ/SMAD signaling pathway and stimulate the development of epilepsy, and the inhibition of LTBP1 expression can control the occurrence of epilepsy with neuroprotection.
Collapse
Affiliation(s)
- Bo Liu
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, 161000, China
| | - Yan Wang
- Department of Neurology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, 161000, China
| | - Dongruo He
- Department of Neurophysiology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, 161000, China
| | - Guochao Han
- Department of Neurophysiology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, 161000, China
| | - Hao Wang
- Department of Neurophysiology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, 161000, China
| | - Yuan Lin
- Department of Neurophysiology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, 161000, China
| | - Tianyu Zhang
- Department of CT, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, 161000, China
| | - Chao Yi
- Department of Neurosurgery, Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, 161000, China
| | - Hui Li
- Department of Neurophysiology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, 161000, China
| |
Collapse
|
11
|
Prakash N. Developmental pathways linked to the vulnerability of adult midbrain dopaminergic neurons to neurodegeneration. Front Mol Neurosci 2022; 15:1071731. [PMID: 36618829 PMCID: PMC9815185 DOI: 10.3389/fnmol.2022.1071731] [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: 10/16/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The degeneration of dopaminergic and other neurons in the aging brain is considered a process starting well beyond the infantile and juvenile period. In contrast to other dopamine-associated neuropsychiatric disorders, such as schizophrenia and drug addiction, typically diagnosed during adolescence or young adulthood and, thus, thought to be rooted in the developing brain, Parkinson's Disease (PD) is rarely viewed as such. However, evidences have accumulated suggesting that several factors might contribute to an increased vulnerability to death of the dopaminergic neurons at an already very early (developmental) phase in life. Despite the remarkable ability of the brain to compensate such dopamine deficits, the early loss or dysfunction of these neurons might predispose an individual to suffer from PD because the critical threshold of dopamine function will be reached much earlier in life, even if the time-course and strength of naturally occurring and age-dependent dopaminergic cell death is not markedly altered in this individual. Several signaling and transcriptional pathways required for the proper embryonic development of the midbrain dopaminergic neurons, which are the most affected in PD, either continue to be active in the adult mammalian midbrain or are reactivated at the transition to adulthood and under neurotoxic conditions. The persistent activity of these pathways often has neuroprotective functions in adult midbrain dopaminergic neurons, whereas the reactivation of silenced pathways under pathological conditions can promote the survival and even regeneration of these neurons in the lesioned or aging brain. This article summarizes our current knowledge about signaling and transcription factors involved in midbrain dopaminergic neuron development, whose reduced gene dosage or signaling activity are implicated in a lower survival rate of these neurons in the postnatal or aging brain. It also discusses the evidences supporting the neuroprotection of the midbrain dopaminergic system after the external supply or ectopic expression of some of these secreted and nuclear factors in the adult and aging brain. Altogether, the timely monitoring and/or correction of these signaling and transcriptional pathways might be a promising approach to a much earlier diagnosis and/or prevention of PD.
Collapse
|
12
|
Selvakumar SC, Preethi KA, Tusubira D, Sekar D. MicroRNAs in the epigenetic regulation of disease progression in Parkinson's disease. Front Cell Neurosci 2022; 16:995997. [PMID: 36187290 PMCID: PMC9524246 DOI: 10.3389/fncel.2022.995997] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 08/31/2022] [Indexed: 12/28/2022] Open
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative condition with symptoms such as resting tremor, rigidity, bradykinesia (slowness of moment), and postural instability. Neuroinflammation plays a significant part in the onset and progression of neurodegeneration in a wide range of disorders, including PD. The loss of dopaminergic neurons in the substantia nigra (SN) is thought to be the primary cause of PD disease progression. However, other neurotransmitter systems like serotoninergic, glutamatergic, noradrenergic, adrenergic, cholinergic, tryptaminergic, and peptidergic appear to be affected as well. Epigenetic regulation of gene expression is emerging as an influencing factor in the pathophysiology of PD. In recent years, epigenetic regulation by microRNAs (miRNAs) has been discovered to play an important function in the disease progression of PD. This review explores the role of miRNAs and their signaling pathways in regulating gene expression from development through neurodegeneration and how these mechanisms are linked to the pathophysiology of PD, emphasizing potential therapeutic interventions.
Collapse
Affiliation(s)
- Sushmaa Chandralekha Selvakumar
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - K. Auxzilia Preethi
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Deusdedit Tusubira
- Department of Biochemistry, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Durairaj Sekar
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
13
|
Protective effect of 3-n-butylphthalide against intrastriatal injection of malonic acid-induced neurotoxicity and biochemical alteration in rats. Biomed Pharmacother 2022; 155:113664. [PMID: 36095961 DOI: 10.1016/j.biopha.2022.113664] [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: 07/04/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 11/20/2022] Open
Abstract
Mitochondrial abnormalities and a defective expression of neurotrophic factors contribute to neuronal damage in Huntington's disease (HD). HD patients showed a reduction in transforming growth factor-β1 (TGF-β1) levels in the peripheral blood and in cortical neurons. 3-n-butylphthalide (NBP) is first isolated from the seeds of celery, treats ischemic stroke in China. NBP could attenuate cognitive and motor impairments in the experimental models of Parkinson's disease and Alzheimer's disease, reduce mitochondrial oxidative stress and increase the expression of TGF-β1 in rats with focal cerebral ischemia. To our knowledge, the effect of NBP on Huntington's disease has not been reported. We proposed the hypothesis that whether NBP could protect mitochondria and regulate TGF-β1 and its downstream signaling in a HD animal model, further prevents motor dysfunction. Malonic acid is a reversible inhibitor of mitochondrial enzyme complex-II, induces energy crisis and free radical generation. In this study, we used intrastriatal injections of malonic acid in rats to mimic mitochondrial abnormalities and the other HD like symptoms. We found that treatment with NBP significantly attenuated malonic acid-induced motor and cognitive dysfunction in locomotor behaviour test, rotarod test, novel object recognition test and morris water maze test, prevented neurotoxicity and mitochondrial damage, activated TGF-β1/Akt/Wnt/β-Catenin pathway in striatum, but didn't regulate mitochondrial fusion and fission. The above effect was partly reversed by a PI3K/Akt inhibitor. Our data support NBP as a potential candidate for the treatment of HD.
Collapse
|
14
|
Liu Q, Huang Y, Duan M, Yang Q, Ren B, Tang F. Microglia as Therapeutic Target for Radiation-Induced Brain Injury. Int J Mol Sci 2022; 23:8286. [PMID: 35955439 PMCID: PMC9368164 DOI: 10.3390/ijms23158286] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Radiation-induced brain injury (RIBI) after radiotherapy has become an increasingly important factor affecting the prognosis of patients with head and neck tumor. With the delivery of high doses of radiation to brain tissue, microglia rapidly transit to a pro-inflammatory phenotype, upregulate phagocytic machinery, and reduce the release of neurotrophic factors. Persistently activated microglia mediate the progression of chronic neuroinflammation, which may inhibit brain neurogenesis leading to the occurrence of neurocognitive disorders at the advanced stage of RIBI. Fully understanding the microglial pathophysiology and cellular and molecular mechanisms after irradiation may facilitate the development of novel therapy by targeting microglia to prevent RIBI and subsequent neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Qun Liu
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Yan Huang
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Mengyun Duan
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China; (M.D.); (Q.Y.)
| | - Qun Yang
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China; (M.D.); (Q.Y.)
| | - Boxu Ren
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
| |
Collapse
|
15
|
Ma R, Kutchy NA, Chen L, Meigs DD, Hu G. Primary cilia and ciliary signaling pathways in aging and age-related brain disorders. Neurobiol Dis 2022; 163:105607. [PMID: 34979259 PMCID: PMC9280856 DOI: 10.1016/j.nbd.2021.105607] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 12/08/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Brain disorders are characterized by the progressive loss of structure and function of the brain as a consequence of progressive degeneration and/or death of nerve cells. Aging is a major risk factor for brain disorders such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and stroke. Various cellular and molecular events have been shown to play a role in the progress of neurodegenerative diseases. Emerging studies suggest that primary cilia could be a key regulator in brain diseases. The primary cilium is a singular cellular organelle expressed on the surface of many cell types, such as astrocytes and neurons in the mature brain. Primary cilia detect extracellular cues, such as Sonic Hedgehog (SHH) protein, and transduce these signals into cells to regulate various signaling pathways. Abnormalities in ciliary length and frequency (ratio of ciliated cells) have been implicated in various human diseases, including brain disorders. This review summarizes current findings and thoughts on the role of primary cilia and ciliary signaling pathways in aging and age-related brain disorders.
Collapse
Affiliation(s)
- Rong Ma
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Naseer A Kutchy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Anatomy, Physiology and Pharmacology, School of Veterinary Medicine, St. George's University, Grenada
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou, Guangdong 515063, China; Key Laboratory of Intelligent Manufacturing Technology, Ministry of Education, Shantou University, Shantou, Guangdong 515063, China
| | - Douglas D Meigs
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| |
Collapse
|
16
|
Huang W, Tao Y, Zhang X, Zhang X. TGF-β1/SMADs signaling involved in alleviating inflammation induced by nanoparticulate titanium dioxide in BV2 cells. Toxicol In Vitro 2022; 80:105303. [PMID: 34990773 DOI: 10.1016/j.tiv.2021.105303] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
Abstract
There are increasing safety concerns accompanying the widespread use of nanoparticulate titanium dioxide (nano-TiO2). It has been demonstrated that nano-TiO2 can cross the blood-brain barrier and enter the brain, causing damage to the nervous system, consisting mainly of neuroinflammation and neuronal apoptosis. Several studies have linked the TGF-β1/SMADs signaling to the development of inflammatory response in various organs. However, no studies have connected the induction of microglial inflammation by nano-TiO2 to this signaling. Therefore, this study aimed to investigate the role of TGF-β1/SMADs signaling in microglia inflammatory response induced by nano-TiO2. The results showed that nano-TiO2 increased the secretions of pro-inflammatory cytokines (IL-1α, IL-6, and TNF-α) and decreased the expressions of TGF-β1 and SMAD1/2/3 proteins in BV2 cells. When TGF-β1/SMADs signaling was inhibited, the inflammatory effect induced by nano-TiO2 increased, suggesting a suppressive effect of this signaling on the inflammation. In addition, exogenous TGF-β1 upregulated the expressions of TGF-β1 and SMADs1/2/3 proteins as well as decreased the secretions of pro-inflammatory cytokines (IL-1α, IL-6, and TNF-α) compared to BV2 cells treated with only nano-TiO2. Our results suggest that nano-TiO2 may inhibit the TGF-β1/SMADs signaling by suppressing the intracellular secretion of active TGF-β1, leading to microglial activation and the induction or exacerbation of inflammatory responses.
Collapse
Affiliation(s)
- Wendi Huang
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yifan Tao
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiuwen Zhang
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiaoqiang Zhang
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
17
|
Kretzschmar F, Piecha R, Jahn J, Potru PS, Spittau B. Characterization of the Leucocyte Immunoglobulin-like Receptor B4 (Lilrb4) Expression in Microglia. BIOLOGY 2021; 10:biology10121300. [PMID: 34943215 PMCID: PMC8698765 DOI: 10.3390/biology10121300] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary In the present study, we provide a detailed characterization of Lilrb4 expression in microglia and peripheral myeloid cells. Our data demonstrate that LILRB4 is a marker for microglia activation, as evidenced by upregulation after lipopolysaccharide treatment and inhibition of microglial TGFβ signaling. Moreover, we provide evidence that microglia express low levels of Lilrb4 in vivo and high levels in vitro, and we clearly demonstrate that LILRB4 is also expressed by bone marrow-derived monocytes and, to a greater extent, by peritoneal macrophages, defining LILRB4 as a surface marker of myeloid cells and not as a microglia-specific marker. Abstract As resident innate immune cells of the CNS, microglia play important essential roles during physiological and pathological situations. Recent reports have described the expression of Lilrb4 in disease-associated and aged microglia. Here, we characterized the expression of Lilrb4 in microglia in vitro and in vivo in comparison with bone marrow-derived monocytes and peritoneal macrophages in mice. Using BV2 cells, primary microglia cultures as well as ex vivo isolated microglia and myeloid cells in combination with qPCR and flow cytometry, we were able to provide a comprehensive characterization of Lilrb4 expression in distinct mouse myeloid cells. Whereas microglia in vivo display low expression of Lilrb4, primary microglia cultures present high levels of surface LILRB4. Among the analyzed peripheral myeloid cells, peritoneal macrophages showed the highest expression levels of Lilrb4. Moreover, LPS treatment and inhibition of microglial TGFβ signaling resulted in significant increases of LILRB4 cell surface levels. Taken together, our data indicate that LILRB4 is a reliable surface marker for activated microglia and further demonstrate that microglial TGFβ signaling is involved in the regulation of Lilrb4 expression during LPS-induced microglia activation.
Collapse
Affiliation(s)
- Felix Kretzschmar
- Institute of Anatomy, Medicine Rostock, University of Rostock, 18055 Rostock, Germany; (F.K.); (R.P.); (J.J.); (P.S.P.)
| | - Robin Piecha
- Institute of Anatomy, Medicine Rostock, University of Rostock, 18055 Rostock, Germany; (F.K.); (R.P.); (J.J.); (P.S.P.)
| | - Jannik Jahn
- Institute of Anatomy, Medicine Rostock, University of Rostock, 18055 Rostock, Germany; (F.K.); (R.P.); (J.J.); (P.S.P.)
| | - Phani Sankar Potru
- Institute of Anatomy, Medicine Rostock, University of Rostock, 18055 Rostock, Germany; (F.K.); (R.P.); (J.J.); (P.S.P.)
- Anatomy and Cell Biology, Medical School OWL, Bielefeld University, 33615 Bielefeld, Germany
| | - Björn Spittau
- Institute of Anatomy, Medicine Rostock, University of Rostock, 18055 Rostock, Germany; (F.K.); (R.P.); (J.J.); (P.S.P.)
- Anatomy and Cell Biology, Medical School OWL, Bielefeld University, 33615 Bielefeld, Germany
- Correspondence:
| |
Collapse
|
18
|
Guan W, Xu DW, Ji CH, Wang CN, Liu Y, Tang WQ, Gu JH, Chen YM, Huang J, Liu JF, Jiang B. Hippocampal miR-206-3p participates in the pathogenesis of depression via regulating the expression of BDNF. Pharmacol Res 2021; 174:105932. [PMID: 34628001 DOI: 10.1016/j.phrs.2021.105932] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022]
Abstract
As a widely-known neuropsychiatric disorder, the exact pathogenesis of depression remains elusive. MiRNA-206 (miR-206) is conventionally known as one of the myomiRs and has two forms: miR-206-3p and miR-206-5p. Recently, miR-206 has been demonstrated to regulate the biosynthesis of brain-derived neurotrophic factor (BDNF), a very popular target involved in depression and antidepressant responses. Here we assumed that miR-206 may play a role in depression, and various methods including the chronic social defeat stress (CSDS) model of depression, quantitative real-time reverse transcription PCR, western blotting, immuofluorescence and virus-mediated gene transfer were used together. It was found that CSDS robustly increased the level of miR-206-3p but not miR-206-5p in the hippocampus. Both genetic overexpression of hippocampal miR-206-3p and intranasal administration of AgomiR-206-3p induced not only notable depressive-like behaviors but also significantly decreased hippocampal BDNF signaling cascade and neurogenesis in naïve C57BL/6J mice. In contrast, both genetic knockdown of hippocampal miR-206-3p and intranasal administration of AntagomiR-206-3p produced significant antidepressant-like effects in the CSDS model of depression. Furthermore, it was found that the antidepressant-like effects induced by miR-206-3p inhibition require the hippocampal BDNF-TrkB system. Taken together, hippocampal miR-206-3p participates in the pathogenesis of depression by regulating BDNF biosynthesis and is a feasible antidepressant target.
Collapse
Affiliation(s)
- Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Da-Wei Xu
- Department of Orthopaedics, Affiliated Hospital 2 of Nantong University, Nantong 226001, Jiangsu, China
| | - Chun-Hui Ji
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Cheng-Niu Wang
- Basic Medical Research Centre, Medical College, Nantong University; Nantong 226001, Jiangsu, China
| | - Yue Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Wen-Qian Tang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Jiang-Hong Gu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Yan-Mei Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Jian-Feng Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
19
|
Xiao Y, Hu X, Fan S, Zhong J, Mo X, Liu X, Hu Y. Single-Cell Transcriptome Profiling Reveals the Suppressive Role of Retinal Neurons in Microglia Activation Under Diabetes Mellitus. Front Cell Dev Biol 2021; 9:680947. [PMID: 34434927 PMCID: PMC8381733 DOI: 10.3389/fcell.2021.680947] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/20/2021] [Indexed: 01/16/2023] Open
Abstract
Diabetic retinopathy, as one of the common complications of diabetes mellitus, is the leading cause of blindness in the working-age population worldwide. The disease is characterized by damage to retinal vasculature, which is associated with the activation of retina microglial and induces chronic neurodegeneration. Previous studies have identified the effects of activated microglial on the retinal neurons, but the cellular and molecular mechanisms underlying microglial activation is largely unknown. Here, we performed scRNA-seq on the retina of non-human primates with diabetes mellitus, and identified cell-type-specific molecular changes of the six major cell types. By identifying the ligand-receptor expression patterns among different cells, we established the interactome of the whole retina. The data showed that TNF-α signal mediated the activation of microglia through an autocrine manner. And we found TGFβ2, which was upregulated in cone dramatically by hyperglycemia, inhibited microglia activation at the early stage of diabetic retinopathy. In summary, our study is the first to profile cell-specific molecular changes and the cell-cell interactome of retina under diabetes mellitus, paving a way to dissect the cellular and molecular mechanisms underlying early-stage diabetic retinopathy.
Collapse
Affiliation(s)
- Yuhua Xiao
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Xing Hu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Shuxin Fan
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Jiawei Zhong
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Xinzhi Mo
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Xialin Liu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Youjin Hu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
20
|
The C-terminal domain of the heavy chain of tetanus toxin prevents the oxidative and nitrosative stress induced by acute toxicity of 1-methyl-4-phenylpyridinium, a rat model of Parkinson's disease. Neurosci Res 2021; 174:36-45. [PMID: 34453989 DOI: 10.1016/j.neures.2021.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/11/2021] [Accepted: 08/22/2021] [Indexed: 12/21/2022]
Abstract
The recombinant carboxyl-terminal domain of the heavy chain of tetanus toxin (Hc-TeTx) exerts neuroprotective and neurorestorative effects on the dopaminergic system of animal models of Parkinson's disease (PD). The present study aimed to determine the effect of the Hc-TeTx fragment on the markers of oxidative stress and nitrosative stress generated by the acute toxicity of 1-methyl-4-phenylpyridinium (MPP+). For this purpose, the Hc-TeTx fragment was administered once a day in three 20 μg/kg consecutive injections into the grastrocnemius muscle of the rats, with an intra-striatal unilateral injection of 1 μL of MPP+ [10 μg/mL] then administered in order to cause a dopaminergic lesion. The results obtained show that the rats treated with Hc-TeTx plus MPP+ presented an increase in the expression of tyrosine hydroxylase (TH), a significantly greater decrease in the levels of the markers of oxidative stress, nitrosative stress, and neurodegeneration than that observed for the group injured with only MPP+. Moreover, it was observed that total superoxide dismutase (SOD) and copper/zinc SOD activity increased with the administration of Hc-TeTx. Finally, immunoreactivity levels were observed to decrease for the levels of 3-nitrotyrosine and the glial fibrillary acidic protein in the ipsilateral striatum of the rats treated with Hc-TeTx plus MPP+, in contrast with those lesioned with MPP+ alone. Our results demonstrate that the recombinant Hc-TeTx fragment may be a potent antioxidant and, therefore, could be suggested as a therapeutic tool against the dopaminergic neuronal impairment observed in the early stages of PD.
Collapse
|
21
|
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
|
22
|
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
|
23
|
Traumatic Brain Injury: Mechanistic Insight on Pathophysiology and Potential Therapeutic Targets. J Mol Neurosci 2021; 71:1725-1742. [PMID: 33956297 DOI: 10.1007/s12031-021-01841-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) causes brain damage, which involves primary and secondary injury mechanisms. Primary injury causes local brain damage, while secondary damage begins with inflammatory activity followed by disruption of the blood-brain barrier (BBB), peripheral blood cells infiltration, brain edema, and the discharge of numerous immune mediators including chemotactic factors and interleukins. TBI alters molecular signaling, cell structures, and functions. Besides tissue damage such as axonal damage, contusions, and hemorrhage, TBI in general interrupts brain physiology including cognition, decision-making, memory, attention, and speech capability. Regardless of the deep understanding of the pathophysiology of TBI, the underlying mechanisms still need to be assessed with a desired therapeutic agent to control the consequences of TBI. The current review gives a brief outline of the pathophysiological mechanism of TBI and various biochemical pathways involved in brain injury, pharmacological treatment approaches, and novel targets for therapy.
Collapse
|
24
|
Jing L, Hou L, Zhang D, Li S, Ruan Z, Zhang X, Hong JS, Wang Q. Microglial Activation Mediates Noradrenergic Locus Coeruleus Neurodegeneration via Complement Receptor 3 in a Rotenone-Induced Parkinson's Disease Mouse Model. J Inflamm Res 2021; 14:1341-1356. [PMID: 33859489 PMCID: PMC8044341 DOI: 10.2147/jir.s299927] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
Background Chronic exposure to the insecticide rotenone can damage dopaminergic neurons and lead to an increased risk of Parkinson’s disease (PD). Whereas it is not clear whether rotenone induces neurodegeneration of noradrenergic locus coeruleus (LC/NE) neurons. Chronic neuroinflammation mediated by microglia has been involved in the pathogenesis of PD. Evidence shows that complement receptor 3 (CR3) is a crucial regulator of microglial activation and related neurodegeneration. However, it is not clear whether CR3 mediates rotenone-elicited degeneration of LC/NE neurons through microglia-mediated neuroinflammation. Materials and Methods Wild type (WT) and CR3 knockout (KO) mice were treated with rotenone. PLX3397 and minocycline were used to deplete or inactivate the microglia. Leukadherin-1 (LA-1) was used to modulate CR3. LC/NE neurodegeneration, microglial phenotype, and expression of CR3 were determined by using immunohistochemistry, Western blot and real-time polymerase chain reaction (PCR) techniques. The glutathione (GSH) and malondialdehyde (MDA) contents were measured by using commercial kits. Results Rotenone exposure led to dose- and time-dependent LC/NE neuronal loss and microglial activation in mice. Depletion of microglia by PLX3397 or inhibition of microglial activation by minocycline significantly reduced rotenone-induced LC/NE neurodegeneration. Mechanistic studies revealed that CR3 played an essential role in the rotenone-induced activation of microglia and neurodegeneration of LC/NE neurons. Rotenone elevated the expression of CR3, and genetic ablation of CR3 markedly reduced rotenone-induced microglial activation and M1 polarization. LA-1 also suppressed rotenone-induced toxic microglial M1 activation. Furthermore, lack of CR3 or treatment with LA-1 reduced oxidative stress in the brainstem of rotenone-intoxicated mice. Finally, we found that mice deficient in CR3 or treated with LA-1 were more resistant to rotenone-induced LC/NE neurodegeneration than WT or vehicle-treated mice, respectively. Conclusion Our results indicate that CR3-mediated microglial activation participates in rotenone-induced LC/NE neurodegeneration, providing novel insight into environmental toxin-induced neurotoxicity and related Parkinsonism.
Collapse
Affiliation(s)
- Lu Jing
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China.,Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Liyan Hou
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Dongdong Zhang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Sheng Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Zhengzheng Ruan
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Xiaomeng Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Qingshan Wang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China.,National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, People's Republic of China
| |
Collapse
|
25
|
Acioglu C, Li L, Elkabes S. Contribution of astrocytes to neuropathology of neurodegenerative diseases. Brain Res 2021; 1758:147291. [PMID: 33516810 DOI: 10.1016/j.brainres.2021.147291] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/10/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
Classically, the loss of vulnerable neuronal populations in neurodegenerative diseases was considered to be the consequence of cell autonomous degeneration of neurons. However, progress in the understanding of glial function, the availability of improved animal models recapitulating the features of the human diseases, and the development of new approaches to derive glia and neurons from induced pluripotent stem cells obtained from patients, provided novel information that altered this view. Current evidence strongly supports the notion that non-cell autonomous mechanisms contribute to the demise of neurons in neurodegenerative disorders, and glia causally participate in the pathogenesis and progression of these diseases. In addition to microglia, astrocytes have emerged as key players in neurodegenerative diseases and will be the focus of the present review. Under the influence of pathological stimuli present in the microenvironment of the diseased CNS, astrocytes undergo morphological, transcriptional, and functional changes and become reactive. Reactive astrocytes are heterogeneous and exhibit neurotoxic (A1) or neuroprotective (A2) phenotypes. In recent years, single-cell or single-nucleus transcriptome analyses unraveled new, disease-specific phenotypes beyond A1/A2. These investigations highlighted the complexity of the astrocytic responses to CNS pathology. The present review will discuss the contribution of astrocytes to neurodegenerative diseases with particular emphasis on Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia. Some of the commonalties and differences in astrocyte-mediated mechanisms that possibly drive the pathogenesis or progression of the diseases will be summarized. The emerging view is that astrocytes are potential new targets for therapeutic interventions. A comprehensive understanding of astrocyte heterogeneity and disease-specific phenotypic complexity could facilitate the design of novel strategies to treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Cigdem Acioglu
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Lun Li
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Stella Elkabes
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| |
Collapse
|
26
|
Domingues AV, Pereira IM, Vilaça-Faria H, Salgado AJ, Rodrigues AJ, Teixeira FG. Glial cells in Parkinson´s disease: protective or deleterious? Cell Mol Life Sci 2020; 77:5171-5188. [PMID: 32617639 PMCID: PMC11104819 DOI: 10.1007/s00018-020-03584-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 05/25/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
Glial cells have been identified more than 100 years ago, and are known to play a key role in the central nervous system (CNS) function. A recent piece of evidence is emerging showing that in addition to the capacity of CNS modulation and homeostasis, glial cells are also being looked like as a promising cell source not only to study CNS pathologies initiation and progression but also to the establishment and development of new therapeutic strategies. Thus, in the present review, we will discuss the current evidence regarding glial cells' contribution to neurodegenerative diseases as Parkinson's disease, providing cellular, molecular, functional, and behavioral data supporting its active role in disease initiation, progression, and treatment. As so, considering their functional relevance, glial cells may be important to the understanding of the underlying mechanisms regarding neuronal-glial networks in neurodegeneration/regeneration processes, which may open new research opportunities for their future use as a target or treatment in human clinical trials.
Collapse
Affiliation(s)
- Ana V Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - Inês M Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - Helena Vilaça-Faria
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - Ana J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal.
| | - Fábio G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.
- ICVS/3B's Associate Lab, PT Government Associated Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
27
|
Xu C, Bai Q, Wang C, Meng Q, Gu Y, Wang Q, Xu W, Han Y, Qin Y, Jia S, Zhang J, Xu J, Li J, Chen M, Wang F. miR-433 Inhibits Neuronal Growth and Promotes Autophagy in Mouse Hippocampal HT-22 Cell Line. Front Pharmacol 2020; 11:536913. [PMID: 33381022 PMCID: PMC7768889 DOI: 10.3389/fphar.2020.536913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 09/30/2020] [Indexed: 11/18/2022] Open
Abstract
Background: MicroRNAs (miRNAs) have an increasing functional role in some neurodegenerative diseases. Autophagy, the degradation of bulk protein in the cytoplasm, is the quality control function of protein and has a protective role in the survival of neural cells. miR-433 may play a regulatory role in neurodegenerative diseases. Many aspects underlying the mechanism of miR-433 in neural development and neurodegeneration are not clear. Methods: In this study, we established stable cell lines expressing miR-433 by infecting mouse hippocampal neural cell line (HT-22) cells with rLV-miR-433 and the control rLV-miR. Pre-miR-433 expression was analyzed using polymerase chain reaction (PCR). Mature miR-433 expression was measured using quantitative PCR (qPCR). The effect of miR-433 overexpression on cell proliferation was determined using a CCK-8 assay and flow cytometry. RNA interference was used to analyze the function of Cdk12 in mediating the effect of miR-433 on cell proliferation. The effect of miR-433 overexpression on cell apoptosis was determined by flow cytometry. Autophagy-related genes Atg4a, LC3B, and Beclin-1 were determined using qPCR, Western blot, or immunofluorescence. In addition, RNA interference was used to analyze the effect of Atg4a on the induction of autophagy. TargetScan 7.2 was used to predict the target genes of miR-433, and Smad9 was determined using qPCR. Results: Our results indicated that miR-433 increased the expression of Atg4a and induced autophagy by increasing the expression of LC3B-Ⅱ and Beclin-1 in an Atg4a-dependent manner. In addition, miR-433 upregulated the expression of Cdk12 and inhibited cell proliferation in a Cdk12-dependent manner and promoted apoptosis in HT-22 cells under the treatment of 10-hydroxycamptothecin. Conclusion: The results of our study suggest that miR-433 may regulate neuronal growth by promoting autophagy and attenuating cell proliferation. This might be a potential therapeutic intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Chunli Xu
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingke Bai
- Department of Neurology, Pudong People's Hospital, Shanghai, China
| | - Chen Wang
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Qiuyu Meng
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Yuming Gu
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiwei Wang
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjie Xu
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Han
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong Qin
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Song Jia
- Teaching Laboratory Center of Medicine and Life Science, Tongji University School of Medicine, Shanghai, China
| | - Junfang Zhang
- Teaching Laboratory Center of Medicine and Life Science, Tongji University School of Medicine, Shanghai, China
| | - Jie Xu
- Teaching Laboratory Center of Medicine and Life Science, Tongji University School of Medicine, Shanghai, China
| | - Jiao Li
- Teaching Laboratory Center of Medicine and Life Science, Tongji University School of Medicine, Shanghai, China
| | - Miao Chen
- Department of Neurology, Shidong hospital, University of Shanghai for Science and Technology, Shanghai, China
| | - Feng Wang
- Department of Neurology, The Seventh People's Hospital of Integrated Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
28
|
Liu Y, Yu L, Xu Y, Tang X, Wang X. Substantia nigra Smad3 signaling deficiency: relevance to aging and Parkinson's disease and roles of microglia, proinflammatory factors, and MAPK. J Neuroinflammation 2020; 17:342. [PMID: 33198771 PMCID: PMC7670688 DOI: 10.1186/s12974-020-02023-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/02/2020] [Indexed: 02/08/2023] Open
Abstract
Background Smad3 signaling is indicated to regulate microglia activity. Parkinson’s disease (PD) neurodegeneration is shown to be associated with aging and neuroinflammation. However, it remains unclear about the relationship among Smad3 signaling, aging, neuroinflammation, and PD. Methods Rats were treated with SIS3 (a specific inhibitor of Smad3, intranigal injection) and/or lipopolysaccharide (intraperitoneal injection). We investigated the effect of SIS3 and lipopolysaccharide and their mechanism of action on motor behavior and nigrostriatal dopaminergic system in the rats. Furthermore, we explored the effect of SIS3 and LPS and their potential signaling mechanism of action on inflammatory response by using primary microglial cultures. Finally, we investigated the relationship among aging, Smad3 signaling, and neuroinflammation using animals of different ages. Results Both SIS3 and lipopolysaccharide induced significant behavior deficits and nigrostriatal dopaminergic neurodegeneration in the rats compared with the vehicle-treated (control) rats. Significantly increased behavior deficits and nigrostriatal dopaminergic neurodegeneration were observed in the rats co-treated with SIS3 and lipopolysaccharide compared with the rats treated with vehicle, SIS3, or lipopolysaccharide. Furthermore, both SIS3 and lipopolysaccharide induced significant microglia activation and proinflammatory factor (IL-1β, IL-6, iNOS, and ROS) level increase in the SN of rats compared with the control rats. Significantly enhanced microglial inflammatory response was observed in the rats co-treated with SIS3 and lipopolysaccharide compared with the other three groups. For our in vitro study, both SIS3 and lipopolysaccharide induced significant proinflammatory factor level increase in primary microglia cultures compared with the control cultures. Significantly increased inflammatory response was observed in the cultures co-treated with SIS3 and lipopolysaccharide compared with the other three groups. MAPK (ERK/p38) contributed to microglial inflammatory response induced by co-treatment with SIS3 and lipopolysaccharide. Interestingly, there was decrease in Smad3 and pSmad3 expression (protein) and enhancement of neuroinflammation in the mouse SN with aging. Proinflammatory factor levels were significantly inversely correlated with Smad3 and pSmad3 expression. Conclusion Our study strongly indicates the involvement of SN Smad3 signaling deficiency in aging and PD neurodegeneration and provides a novel molecular mechanism underlying the participation of aging in PD and helps to elucidate the mechanisms for the combined effect of multiple factors in PD.
Collapse
Affiliation(s)
- Ying Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, P.R. China
| | - Lijia Yu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, P.R. China
| | - Yaling Xu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, P.R. China
| | - Xiaohui Tang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, P.R. China
| | - Xijin Wang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, P.R. China.
| |
Collapse
|
29
|
Najib NH, Nies YH, Abd Halim SA, Yahaya MF, Das S, Lim WL, Teoh SL. Modeling Parkinson’s Disease in Zebrafish. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:386-399. [DOI: 10.2174/1871527319666200708124117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 05/10/2020] [Accepted: 06/17/2020] [Indexed: 01/04/2023]
Abstract
Parkinson’s Disease (PD) is one of the most common neurodegenerative disorders that affects
the motor system, and includes cardinal motor symptoms such as resting tremor, cogwheel rigidity,
bradykinesia and postural instability. Its prevalence is increasing worldwide due to the increase in
life span. Although, two centuries since the first description of the disease, no proper cure with regard
to treatment strategies and control of symptoms could be reached. One of the major challenges faced
by the researchers is to have a suitable research model. Rodents are the most common PD models
used, but no single model can replicate the true nature of PD. In this review, we aim to discuss another
animal model, the zebrafish (Danio rerio), which is gaining popularity. Zebrafish brain has all the major
structures found in the mammalian brain, with neurotransmitter systems, and it also possesses a
functional blood-brain barrier similar to humans. From the perspective of PD research, the zebrafish
possesses the ventral diencephalon, which is thought to be homologous to the mammalian substantia
nigra. We summarize the various zebrafish models available to study PD, namely chemical-induced
and genetic models. The zebrafish can complement the use of other animal models for the mechanistic
study of PD and help in the screening of new potential therapeutic compounds.
Collapse
Affiliation(s)
- Nor H.M. Najib
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Yong H. Nies
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Syarifah A.S. Abd Halim
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Mohamad F. Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Srijit Das
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Wei L. Lim
- Department of Biological Sciences, School of Science and Technology, Sunway University, Selangor, Malaysia
| | - Seong L. Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
30
|
Islas-Cortez M, Rios C, Rubio-Osornio M, Zamudio S, Orozco-Suarez S, Mendez-Armenta M, Nava-Ruiz C, Diaz-Ruiz A. Characterization of the antiapoptotic effect of copper sulfate on striatal and midbrain damage induced by MPP + in rats. Neurotoxicology 2020; 82:18-25. [PMID: 33127410 DOI: 10.1016/j.neuro.2020.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/18/2023]
Abstract
1-Methyl-4-phenylpyridinium ion (MPP+)-induced neurotoxicity produces cellular damage resembling that encountered in Parkinson's disease. The mechanisms of cellular death after MPP+ include the participation of oxidative stress in the loss of dopaminergic neurons. Among the mechanisms of defense against oxidative stress, several copper-dependent proteins have been implicated: Cu/Zn-SOD, ceruloplasmin, and metallothionein. Another important mechanism of damage, is MPP + interference with mitochondrial respiration. Both, oxidative stress and inhibition of mitochondrial respiration may trigger apoptosis in the neurons after MPP+. The aim of the present study was to characterize the time-course of apoptosis induced by MPP+ to determine if copper sulfate pretreatment is able to prevent the activation of caspases and decreased the neuronal apoptosis. MPP+ was microinjected into rat striatum using a stereotactic frame. The results showed increased activities of caspases 8, 9 and 3, between 72-120 hours after administration of MPP+, both in striatum and midbrain. After this study, we tested the effect of CuSO4 on MPP+ neurotoxicity, showing a diminution of the apoptotic damage induced by MPP+, decreased levels of enzymatic activity of caspases: 8 (-34 and -25 %), 9 (-25 and -42 %) and 3 (-40 and -29 %) in striatum and midbrain, respectively. Finally, we performed an immunohistochemical analysis, evidencing a decreased number of apoptotic cells in the groups pretreated with copper sulfate pretreatment compared to the control group. With these findings, it is concluded that pretreatment with copper sulfate may be a good alternative to prevent MPP+-induced apoptosis.
Collapse
Affiliation(s)
- Marcela Islas-Cortez
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico; Departamento de Fisiología, Laboratorio de Neurociencia Conductual, Instituto Politécnico Nacional, Unidad Zacatenco, Ciudad de México, Mexico
| | - Camilo Rios
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico; Laboratorio de Neurofarmacología Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México, Mexico
| | - Moisés Rubio-Osornio
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico
| | - Sergio Zamudio
- Departamento de Fisiología, Laboratorio de Neurociencia Conductual, Instituto Politécnico Nacional, Unidad Zacatenco, Ciudad de México, Mexico
| | - Sandra Orozco-Suarez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Ciudad de México, Mexico
| | - Marisela Mendez-Armenta
- Departamento de Neuropatología Experimental Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico
| | - Concepción Nava-Ruiz
- Departamento de Neuropatología Experimental Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico
| | - Araceli Diaz-Ruiz
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Ciudad de México, Mexico.
| |
Collapse
|
31
|
Prasad EM, Hung SY. Behavioral Tests in Neurotoxin-Induced Animal Models of Parkinson's Disease. Antioxidants (Basel) 2020; 9:E1007. [PMID: 33081318 PMCID: PMC7602991 DOI: 10.3390/antiox9101007] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Currently, neurodegenerative diseases are a major cause of disability around the world. Parkinson's disease (PD) is the second-leading cause of neurodegenerative disorder after Alzheimer's disease. In PD, continuous loss of dopaminergic neurons in the substantia nigra causes dopamine depletion in the striatum, promotes the primary motor symptoms of resting tremor, bradykinesia, muscle rigidity, and postural instability. The risk factors of PD comprise environmental toxins, drugs, pesticides, brain microtrauma, focal cerebrovascular injury, aging, and hereditary defects. The pathologic features of PD include impaired protein homeostasis, mitochondrial dysfunction, nitric oxide, and neuroinflammation, but the interaction of these factors contributing to PD is not fully understood. In neurotoxin-induced PD models, neurotoxins, for instance, 6-hydroxydopamine (6-OHDA), 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 1-Methyl-4-phenylpyridinium (MPP+), paraquat, rotenone, and permethrin mainly impair the mitochondrial respiratory chain, activate microglia, and generate reactive oxygen species to induce autooxidation and dopaminergic neuronal apoptosis. Since no current treatment can cure PD, using a suitable PD animal model to evaluate PD motor symptoms' treatment efficacy and identify therapeutic targets and drugs are still needed. Hence, the present review focuses on the latest scientific developments in different neurotoxin-induced PD animal models with their mechanisms of pathogenesis and evaluation methods of PD motor symptoms.
Collapse
Affiliation(s)
- E. Maruthi Prasad
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, No.91, Hsueh-Shih Road, Taichung 40402, Taiwan;
| | - Shih-Ya Hung
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, No.91, Hsueh-Shih Road, Taichung 40402, Taiwan;
- Department of Medical Research, China Medical University Hospital, No. 2, Yude Road, Taichung 40447, Taiwan
| |
Collapse
|
32
|
Quarta A, Meese T, Pieters Z, Van Breedam E, Le Blon D, Van Broeckhoven J, Hendrix S, Goossens H, Hens N, Berneman Z, Van Nieuwerburgh F, Ponsaerts P. Murine induced pluripotent stem cell-derived neuroimmune cell culture models emphasize opposite immune-effector functions of interleukin 13-primed microglia and macrophages in terms of neuroimmune toxicity. Glia 2020; 69:326-345. [PMID: 32865285 DOI: 10.1002/glia.23899] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/19/2022]
Abstract
Cellular models of induced pluripotent stem cell (iPSC)-derived microglia and macrophages are an emerging toolbox to investigate neuroinflammation in vitro. We previously demonstrated that murine iPSC-microglia and iPSC-macrophages display phenotypical activation properties highly comparable to microglia and macrophages in vivo. Here we extended the characterization of iPSC-microglia and iPSC-macrophages with the analysis of their transcriptome profile. Next, these cellular models were employed to evaluate neuroimmune toxicity in vitro and to investigate the immune-modulatory properties of interleukin 13 (IL13), a cytokine known for its ability to protect against neuroinflammation-induced pathology by modulating microglia and macrophage activation. iPSC-microglia and iPSC-macrophages, in co-culture with astrocyte-committed neural stem cells (NSC), were (pre)treated with IL13 and stimulated with lipopolysaccharide (LPS) and interferon γ (IFNγ), to assess how IL13 modulates their inflammatory response. Additionally, the use of luciferase-expressing NSC (Luc-NSC) allowed real-time monitoring of immune-mediated neurotoxicity. Despite the known anti-inflammatory properties of IL13, iPSC-microglia primed with IL13 before LPS + IFNγ stimulation significantly increased NO secretion. This was associated with a marked reduction of the luminescence signal produced by Luc-NSC. Interestingly, we observed that IL13 signaling has a divergent functional outcome in microglia as compared to macrophages, as for the latter no major alterations in NO release and Luc-NSC viability were observed upon IL13 (pre)treatment. Finally, the striking IL13-induced upregulation of NO secretion by microglia under pro-inflammatory conditions was confirmed in vivo, where intracerebral delivery of IL13 increased inducible nitric oxide synthase mRNA expression. Concluding, we applied iPSC-derived neuroimmune cell culture models to identify distinct neuroimmune (toxicity) responses of microglia and macrophages to IL13-based immune modulation.
Collapse
Affiliation(s)
- Alessandra Quarta
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Tim Meese
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Zoë Pieters
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.,Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), Data Science Institute, Hasselt University, Hasselt, Belgium.,Centre for Health Economics Research and Modelling Infectious Diseases, University of Antwerp, Antwerp, Belgium
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Debbie Le Blon
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sven Hendrix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Niel Hens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.,Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), Data Science Institute, Hasselt University, Hasselt, Belgium.,Centre for Health Economics Research and Modelling Infectious Diseases, University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
33
|
Fatoba O, Itokazu T, Yamashita T. Microglia as therapeutic target in central nervous system disorders. J Pharmacol Sci 2020; 144:102-118. [PMID: 32921391 DOI: 10.1016/j.jphs.2020.07.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/19/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic microglial activation is associated with the pathogenesis of several CNS disorders. Microglia show phenotypic diversity and functional complexity in diseased CNS. Thus, understanding the pathology-specific heterogeneity of microglial behavior is crucial for the future development of microglia-modulating therapy for variety of CNS disorders. This review summarizes up-to-date knowledge on how microglia contribute to CNS homeostasis during development and throughout adulthood. We discuss the heterogeneity of microglial phenotypes in the context of CNS disorders with an emphasis on neurodegenerative diseases, demyelinating diseases, CNS trauma, and epilepsy. We conclude this review with a discussion about the disease-specific heterogeneity of microglial function and how it could be exploited for therapeutic intervention.
Collapse
Affiliation(s)
- Oluwaseun Fatoba
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
34
|
The Role of TGFβ Signaling in Microglia Maturation and Activation. Trends Immunol 2020; 41:836-848. [PMID: 32741652 DOI: 10.1016/j.it.2020.07.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/23/2022]
Abstract
The pleiotropic cytokine transforming growth factor-beta 1 (TGFβ1) plays pivotal roles in different cell types, including immune cells such as T cells, monocytes/macrophages, and microglia. Microglia are essential during physiological and pathological events. Maturation of postnatal microglia, as well as the regulation of the complex functional repertoire of microglia, needs to be carefully orchestrated. However, an understanding of how mammalian microglia maturation and disease-associated microglia activation is regulated remains fragmentary. Here, we summarize recent observations made by employing transgenic approaches to silence microglial TGFβ signaling in mice. These revealed that TGFβ1 and TGFβ signaling are indispensable for microglia maturation, adult microglia homeostasis, and the control of microglia activation in central nervous system pathologies.
Collapse
|
35
|
Apamin Suppresses LPS-Induced Neuroinflammatory Responses by Regulating SK Channels and TLR4-Mediated Signaling Pathways. Int J Mol Sci 2020; 21:ijms21124319. [PMID: 32560481 PMCID: PMC7352249 DOI: 10.3390/ijms21124319] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/11/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation plays a vital role in neurodegenerative conditions. Microglia are a key component of the neuroinflammatory response. There is a growing interest in developing drugs to target microglia and thereby control neuroinflammatory processes. Apamin (APM) is a specifically selective antagonist of small conductance calcium-activated potassium (SK) channels. However, its effect on neuroinflammation is largely unknown. We examine the effects of APM on lipopolysaccharide (LPS)-stimulated BV2 and rat primary microglial cells. Regarding the molecular mechanism by which APM significantly inhibits proinflammatory cytokine production and microglial cell activation, we found that APM does so by reducing the expression of phosphorylated CaMKII and toll-like receptor (TLR4). In particular, APM potently suppressed the translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/signal transducer and activator of transcription (STAT)3 and phosphorylated mitogen-activated protein kinases (MAPK)-extracellular signal-regulated kinase (ERK). In addition, the correlation of NF-κB/STAT3 and MAPK-ERK in the neuroinflammatory response was verified through inhibitors. The literature and our findings suggest that APM is a promising candidate for an anti-neuroinflammatory agent and can potentially be used for the prevention and treatment of various neurological disorders.
Collapse
|
36
|
Jiang W, Wang X, Wang W, Hua F, Zhang Z, Zhang Z, Xiang J, Yang X. Inhibition of NK1R attenuates LPS-induced microglial inflammation and consequent death of PC12 cells. Brain Res Bull 2020; 162:115-124. [PMID: 32540418 DOI: 10.1016/j.brainresbull.2020.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 10/24/2022]
Abstract
Microglia, the resident immune cells in the central nervous system, play a critical role under physiological conditions, but they may be activated and exaggerate the pathological development of Parkinson's disease (PD). Recent reports have suggested that neurokinin 1 receptor (NK1R) is involved in various inflammatory diseases, including PD. However, whether neurokinin 1 (NK1) is involved in the activation of microglial cells remains unclear. In the present study, we found that (1) NK1R is located in microglial cells and upregulated in lipopolysaccharide (LPS)-activated BV2 microglia. Application of CP-99994, a selective antagonist of NK1R, inhibited the production of inflammatory mediators such as tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), IL-6, inducible macrophage-type nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) in activated BV2 cells. (2) NK1R antagonist suppressed the morphological changes in LPS-stimulated BV2. (3) Microglial inactivation by NK1R antagonist resulted in decreased microglial migration. (4) NK1R antagonist reduced nuclear translocation of nuclear factor kappa-B (NF-κB) and attenuated phosphorylation of mitogen-activated protein kinases (MAPKs) in LPS-stimulated BV2. (5) The cell death of PC12 induced by microglia-mediated neuroinflammation was reversed in a Transwell co-culture system by NK1R antagonist. Collectively, these results showed that inhibition of NK1R attenuates LPS-induced microglial inflammatory response and dopaminergic neurotoxicity, which may be due to the decreased MAPK/NF-κB signal pathway. Thus, NK1R may be a therapeutic target in neuroinflammation, especially in PD.
Collapse
Affiliation(s)
- Weifeng Jiang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiaoying Wang
- Department of Ultrasound, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Wei Wang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zunsheng Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zuohui Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Jie Xiang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Xinxin Yang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| |
Collapse
|
37
|
Wallings RL, Herrick MK, Tansey MG. LRRK2 at the Interface Between Peripheral and Central Immune Function in Parkinson's. Front Neurosci 2020; 14:443. [PMID: 32508566 PMCID: PMC7253584 DOI: 10.3389/fnins.2020.00443] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/09/2020] [Indexed: 12/20/2022] Open
Abstract
It is becoming increasingly accepted that there is an interplay between the peripheral immune response and neuroinflammation in the pathophysiology of Parkinson's disease (PD). Mutations in the leucine-rich-repeat kinase 2 (LRRK2) gene are associated with familial and sporadic cases of PD but are also found in immune-related disorders, such as inflammatory bowel disease (IBD) and leprosy. Furthermore, LRRK2 has been associated with bacterial infections such as Mycobacterium tuberculosis and Salmonella typhimurium. Recent evidence suggests a role of LRRK2 in the regulation of the immune system and modulation of inflammatory responses, at a systemic level, with LRRK2 functionally implicated in both the immune system of the central nervous system (CNS) and the periphery. It has therefore been suggested that peripheral immune signaling may play an important role in the regulation of neurodegeneration in LRRK2 as well as non-LRRK2-associated PD. This review will discuss the current evidence for this hypothesis and will provide compelling rationale for placing LRRK2 at the interface between peripheral immune responses and neuroinflammation.
Collapse
Affiliation(s)
- Rebecca L. Wallings
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
| | - Mary K. Herrick
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
- Laney Graduate School, Emory University, Atlanta, GA, United States
| | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
38
|
Li C, Wu X, Liu S, Shen D, Zhu J, Liu K. Role of Resolvins in the Inflammatory Resolution of Neurological Diseases. Front Pharmacol 2020; 11:612. [PMID: 32457616 PMCID: PMC7225325 DOI: 10.3389/fphar.2020.00612] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
The occurrence of neurological diseases including neurodegenerative disorders, neuroimmune diseases, and cerebrovascular disorders is closely related to neuroinflammation. Inflammation is a response against infection or injury. Genetic abnormalities, the aging process, or environmental factors can lead to dysregulation of the inflammatory response. Our immune system can cause massive damage when the inflammatory response becomes dysregulated. Inflammatory resolution is an effective process that terminates the inflammatory response to maintain health. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are omega-three polyunsaturated fatty acids that play a crucial regulatory role in the development of inflammation. Resolvins (Rvs) derived from EPA and DHA constitute the Rvs E and Rvs D series, respectively. Numerous studies on the effect of Rvs over inflammation using animal models reveal that they have both anti-inflammatory and pro-resolving capabilities. Here, we review the current knowledge on the classification, biosynthesis, receptors, mechanisms of action, and role of Rvs in neurological diseases.
Collapse
Affiliation(s)
- Chunrong Li
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiujuan Wu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Shan Liu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Donghui Shen
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China.,Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Kangding Liu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
39
|
Cao BB, Zhang XX, Du CY, Liu Z, Qiu YH, Peng YP. TGF-β1 Provides Neuroprotection via Inhibition of Microglial Activation in 3-Acetylpyridine-Induced Cerebellar Ataxia Model Rats. Front Neurosci 2020; 14:187. [PMID: 32265625 PMCID: PMC7099147 DOI: 10.3389/fnins.2020.00187] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/20/2020] [Indexed: 12/19/2022] Open
Abstract
Cerebellar ataxias (CAs) consist of a heterogeneous group of neurodegenerative diseases hallmarked by motor deficits and deterioration of the cerebellum and its associated circuitries. Neuroinflammatory responses are present in CA brain, but how neuroinflammation may contribute to CA pathogenesis remain unresolved. Here, we investigate whether transforming growth factor (TGF)-β1, which possesses anti-inflammatory and neuroprotective properties, can ameliorate the microglia-mediated neuroinflammation and thereby alleviate neurodegeneration in CA. In the current study, we administered TGF-β1 via the intracerebroventricle (ICV) in CA model rats, by intraperitoneal injection of 3-acetylpyridine (3-AP), to reveal the neuroprotective role of TGF-β1. The TGF-β1 administration after 3-AP injection ameliorated motor impairments and reduced the calbindin-positive neuron loss and apoptosis in the brain stem and cerebellum. Meanwhile, 3-AP induced microglial activation and inflammatory responses in vivo, which were determined by morphological alteration and an increase in expression of CD11b, enhancement of percentage of CD40 + and CD86 + microglial cells, upregulation of pro-inflammatory mediators, tumor necrosis factor (TNF)-α and interleukin (IL)-1β, and a downregulation of neurotrophic factor, insulin-like growth factor (IGF)-1 in the brain stem and cerebellum. TGF-β1 treatment significantly prevented all the changes caused by 3-AP. In addition, in vitro experiments, TGF-β1 directly attenuated 3-AP-induced microglial activation and inflammatory responses in primary cultures. Purkinje cell exposure to supernatants of primary microglia that had been treated with TGF-β1 reduced neuronal loss and apoptosis induced by 3-AP-treated microglial supernatants. Furthermore, the protective effect was similar to those treated with TNF-α-neutralizing antibody. These findings suggest that TGF-β1 protects against neurodegeneration in 3-AP-induced CA rats via inhibiting microglial activation and at least partly TNF-α release.
Collapse
Affiliation(s)
- Bei-Bei Cao
- Department of Physiology, School of Medicine, Nantong University, Nantong, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiao-Xian Zhang
- Department of Physiology, School of Medicine, Nantong University, Nantong, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chen-Yu Du
- Department of Physiology, School of Medicine, Nantong University, Nantong, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zhan Liu
- Department of Physiology, School of Medicine, Nantong University, Nantong, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, Nantong University, Nantong, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, Nantong University, Nantong, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
40
|
Dong X, Tan NB, Howell KB, Barresi S, Freeman JL, Vecchio D, Piccione M, Radio FC, Calame D, Zong S, Eggers S, Scheffer IE, Tan TY, Van Bergen NJ, Tartaglia M, Christodoulou J, White SM. Bi-allelic LoF NRROS Variants Impairing Active TGF-β1 Delivery Cause a Severe Infantile-Onset Neurodegenerative Condition with Intracranial Calcification. Am J Hum Genet 2020; 106:559-569. [PMID: 32197075 PMCID: PMC7118692 DOI: 10.1016/j.ajhg.2020.02.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 02/26/2020] [Indexed: 01/05/2023] Open
Abstract
Negative regulator of reactive oxygen species (NRROS) is a leucine-rich repeat-containing protein that uniquely associates with latent transforming growth factor beta-1 (TGF- β1) and anchors it on the cell surface; this anchoring is required for activation of TGF-β1 in macrophages and microglia. We report six individuals from four families with bi-allelic variants in NRROS. All affected individuals had neurodegenerative disease with refractory epilepsy, developmental regression, and reduced white matter volume with delayed myelination. The clinical course in affected individuals began with normal development or mild developmental delay, and the onset of seizures occurred within the first year of life, followed by developmental regression. Intracranial calcification was detected in three individuals. The phenotypic features in affected individuals are consistent with those observed in the Nrros knockout mouse, and they overlap with those seen in the human condition associated with TGF-β1 deficiency. The disease-causing NRROS variants involve two significant functional NRROS domains. These variants result in aberrant NRROS proteins with impaired ability to anchor latent TGF-β1 on the cell surface. Using confocal microscopy in HEK293T cells, we demonstrate that wild-type and mutant NRROS proteins co-localize with latent TGF-β1 intracellularly. However, using flow cytometry, we show that our mutant NRROS proteins fail to anchor latent TGF-β1 at the cell surface in comparison to wild-type NRROS. Moreover, wild-type NRROS rescues the defect of our disease-associated mutants in presenting latent TGF-β1 to the cell surface. Taken together, our findings suggest that loss of NRROS function causes a severe childhood-onset neurodegenerative condition with features suggestive of a disordered response to inflammation.
Collapse
Affiliation(s)
- Xiaomin Dong
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Natalie B Tan
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia; Victorian Clinical Genetics Services, Parkville, Victoria 3052, Australia
| | - Katherine B Howell
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia; Department of Neurology, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Sabina Barresi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - Jeremy L Freeman
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Department of Neurology, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Davide Vecchio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - Maria Piccione
- Department of Science for Health Promotion and Mother and Child Care, Università degli Studi di Palermo, Palermo 90127, Italy
| | | | - Daniel Calame
- Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| | - Shan Zong
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
| | - Stefanie Eggers
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Victorian Clinical Genetics Services, Parkville, Victoria 3052, Australia
| | - Ingrid E Scheffer
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia; Department of Neurology, Royal Children's Hospital, Parkville, Victoria 3052, Australia; Department of Medicine, University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Tiong Y Tan
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia; Victorian Clinical Genetics Services, Parkville, Victoria 3052, Australia
| | - Nicole J Van Bergen
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - John Christodoulou
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia; Victorian Clinical Genetics Services, Parkville, Victoria 3052, Australia.
| | - Susan M White
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia; Victorian Clinical Genetics Services, Parkville, Victoria 3052, Australia.
| |
Collapse
|
41
|
Essential role of microglial transforming growth factor-β1 in antidepressant actions of (R)-ketamine and the novel antidepressant TGF-β1. Transl Psychiatry 2020; 10:32. [PMID: 32066676 PMCID: PMC7026089 DOI: 10.1038/s41398-020-0733-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/02/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022] Open
Abstract
In rodent models of depression, (R)-ketamine has greater potency and longer-lasting antidepressant effects than (S)-ketamine; however, the precise molecular mechanisms underlying the antidepressant actions of (R)-ketamine remain unknown. Using RNA-sequencing analysis, we identified novel molecular targets that contribute to the different antidepressant effects of the two enantiomers. Either (R)-ketamine (10 mg/kg) or (S)-ketamine (10 mg/kg) was administered to susceptible mice after chronic social defeat stress (CSDS). RNA-sequencing analysis of prefrontal cortex (PFC) and subsequent GSEA (gene set enrichment analysis) revealed that transforming growth factor (TGF)-β signaling might contribute to the different antidepressant effects of the two enantiomers. (R)-ketamine, but not (S)-ketamine, ameliorated the reduced expressions of Tgfb1 and its receptors (Tgfbr1 and Tgfbr2) in the PFC and hippocampus of CSDS susceptible mice. Either pharmacological inhibitors (i.e., RepSox and SB431542) or neutralizing antibody of TGF-β1 blocked the antidepressant effects of (R)-ketamine in CSDS susceptible mice. Moreover, depletion of microglia by the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX3397 blocked the antidepressant effects of (R)-ketamine in CSDS susceptible mice. Similar to (R)-ketamine, the recombinant TGF-β1 elicited rapid and long-lasting antidepressant effects in animal models of depression. Our data implicate a novel microglial TGF-β1-dependent mechanism underlying the antidepressant effects of (R)-ketamine in rodents with depression-like phenotype. Moreover, TGF-β1 and its receptor agonists would likely constitute a novel rapid-acting and sustained antidepressant in humans.
Collapse
|
42
|
Szymura J, Kubica J, Wiecek M, Pera J. The Immunomodulary Effects of Systematic Exercise in Older Adults and People with Parkinson's Disease. J Clin Med 2020; 9:jcm9010184. [PMID: 31936624 PMCID: PMC7019419 DOI: 10.3390/jcm9010184] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/05/2020] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
We sought to investigate whether regular balance training of moderate intensity (BT) has an effect on changes in selected cytokines, neurotrophic factors, CD200 and fractalkine in healthy older adults and participants with Parkinson’s disease (PD). Sixty-two subjects were divided into groups depending on experimental intervention: (1) group of people with PD participating in BT (PDBT), (2) group of healthy older people participating in BT (HBT), (3,4) control groups including healthy individuals (HNT) and people with PD (PDNT). Blood samples were collected twice: before and after 12 weeks of balance exercise (PDBT, HBT), or 12 weeks apart (PDNT, HNT). The study revealed significant increase of interleukin10 (PDBT, p = 0.026; HBT, p = 0.011), β-nerve growth factor (HBT, p = 0.002; PDBT, p = 0.016), transforming growth factor-β1 (PDBT, p = 0.018; HBT, p < 0.004), brain-derived neurotrophic factor (PDBT, p = 0.011; HBT, p < 0.001) and fractalkine (PDBT, p = 0.045; HBT, p < 0.003) concentration only in training groups. In PDBT, we have found a significant decrease of tumor necrosis factor alpha. No training effect on concentration of interleukin6, insulin-like growth factor 1 and CD200 was observed in both training and control groups. Regular training can modulate level of inflammatory markers and induce neuroprotective mechanism to reduce the inflammatory response.
Collapse
Affiliation(s)
- Jadwiga Szymura
- Department of Clinical Rehabilitation, Faculty of Motor Rehabilitation, University of Physical Education in Krakow, 31–571 Krakow, Poland
- Correspondence: (J.S.); (J.K.)
| | - Jadwiga Kubica
- Institute of Physiotherapy, Faculty of Health Science, Jagiellonian University Medical College, 31–126 Krakow, Poland
- Correspondence: (J.S.); (J.K.)
| | - Magdalena Wiecek
- Department of Physiology and Biochemistry, Faculty of Physical Education and Sport, University of Physical Education in Krakow, 31–571 Krakow, Poland;
| | - Joanna Pera
- Department of Neurology, Faculty of Medicine, Jagiellonian University Medical College, 31–503 Krakow, Poland;
| |
Collapse
|
43
|
Liu Z, Qiu AW, Huang Y, Yang Y, Chen JN, Gu TT, Cao BB, Qiu YH, Peng YP. IL-17A exacerbates neuroinflammation and neurodegeneration by activating microglia in rodent models of Parkinson's disease. Brain Behav Immun 2019; 81:630-645. [PMID: 31351185 DOI: 10.1016/j.bbi.2019.07.026] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/17/2019] [Accepted: 07/23/2019] [Indexed: 12/26/2022] Open
Abstract
Neuroinflammation has been involved in pathogenesis of Parkinson's disease (PD), a chronic neurodegenerative disease characterized neuropathologically by progressive dopaminergic neuronal loss in the substantia nigra (SN). We recently have shown that helper T (Th)17 cells facilitate dopaminergic neuronal loss in vitro. Herein, we demonstrated that interleukin (IL)-17A, a proinflammatory cytokine produced mainly by Th17 cells, contributed to PD pathogenesis depending on microglia. Mouse and rat models for PD were prepared by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or striatal injection of 1-methyl-4-phenylpyridinium (MPP+), respectively. Both in MPTP-treated mice and MPP+-treated rats, blood-brain barrier (BBB) was disrupted and IL-17A level increased in the SN but not in cortex. Effector T (Teff) cells that were adoptively transferred via tail veins infiltrated into the brain of PD mice but not into that of normal mice. The Teff cell transfer aggravated nigrostriatal dopaminergic neurodegeneration, microglial activation and motor impairment. Contrarily, IL-17A deficiency alleviated BBB disruption, dopaminergic neurodegeneration, microglial activation and motor impairment. Anti-IL-17A-neutralizing antibody that was injected into lateral cerebral ventricle in PD rats ameliorated the manifestations mentioned above. IL-17A activated microglia but did not directly affect dopaminergic neuronal survival in vitro. IL-17A exacerbated dopaminergic neuronal loss only in the presence of microglia, and silencing IL-17A receptor gene in microglia abolished the IL-17A effect. IL-17A-treated microglial medium that contained higher concentration of tumor necrosis factor (TNF)-α facilitated dopaminergic neuronal death. Further, TNF-α-neutralizing antibody attenuated MPP+-induced neurotoxicity. The findings suggest that IL-17A accelerates neurodegeneration in PD depending on microglial activation and at least partly TNF-α release.
Collapse
Affiliation(s)
- Zhan Liu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ao-Wang Qiu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Yan Huang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ya Yang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jin-Na Chen
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ting-Ting Gu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Bei-Bei Cao
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
44
|
Role of TGF-Beta1/SMAD2/3 Pathway in Retinal Outer Deep Vascular Plexus and Photoreceptor Damage in Rat 50/10 Oxygen-Induced Retinopathy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4072319. [PMID: 31240212 PMCID: PMC6556365 DOI: 10.1155/2019/4072319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/28/2019] [Accepted: 04/28/2019] [Indexed: 11/25/2022]
Abstract
In retinopathy of prematurity (ROP), outer deep vascular plexus (oDVP) was the emerging field, and the mechanisms of photoreceptor dysfunction remained to be explored. ODVP and photoreceptors were related, with oDVP being part of the supplier of oxygen and nutrients to photoreceptors, while their possible relationship in ROP was not clear. TGF-beta1 has been reported indispensable in oDVP development and altered in ROP patients and animal models. We hypothesized that the TGF-beta1 alteration in rat 50/10 oxygen-induced retinopathy (OIR) model contributed to oDVP malformation and exerted consequent effects on photoreceptor development. We first explored the profile of oDVP development in rat after birth and compared the expression of TGF-beta1 and pSMAD2/3 in Normoxia and OIR groups. Afterwards, the inhibitor of the pathway, LY364947, was used to establish the OIR, OIR+LY364947, Normoxia, and Normoxia+LY364947 groups. The oDVP and photoreceptor were examined by Isolectin B4 staining, western-blot of CD31 and Rho, and electron microscopy. ODVP sprouted at postnatal day 10 (D10) and reached the edge of retina at D14. The TGF-beta1/SMAD2/3 pathway was compromised during the critical period of oDVP development. The inhibitor simulated the oDVP retardation, pericyte, and photoreceptor malformation in the Normoxia+LY364947 group and might further compromise the development of oDVP and photoreceptor in the OIR+LY364947 group. The inhibition of the TGF-beta1/SMAD2/3 pathway indicated its critical role in oDVP malformation and photoreceptor damage, suggesting a possible therapeutic target of ROP treatment.
Collapse
|
45
|
Booth HDE, Wessely F, Connor-Robson N, Rinaldi F, Vowles J, Browne C, Evetts SG, Hu MT, Cowley SA, Webber C, Wade-Martins R. RNA sequencing reveals MMP2 and TGFB1 downregulation in LRRK2 G2019S Parkinson's iPSC-derived astrocytes. Neurobiol Dis 2019; 129:56-66. [PMID: 31085228 DOI: 10.1016/j.nbd.2019.05.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 04/09/2019] [Accepted: 05/10/2019] [Indexed: 12/26/2022] Open
Abstract
Non-neuronal cell types such as astrocytes can contribute to Parkinson's disease (PD) pathology. The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is one of the most common known causes of familial PD. To characterize its effect on astrocytes, we developed a protocol to produce midbrain-patterned astrocytes from human induced pluripotent stem cells (iPSCs) derived from PD LRRK2 G2019S patients and healthy controls. RNA sequencing analysis revealed the downregulation of genes involved in the extracellular matrix in PD cases. In particular, transforming growth factor beta 1 (TGFB1), which has been shown to inhibit microglial inflammatory response in a rat model of PD, and matrix metallopeptidase 2 (MMP2), which has been shown to degrade α-synuclein aggregates, were found to be down-regulated in LRRK2 G2019S astrocytes. Our findings suggest that midbrain astrocytes carrying the LRRK2 G2019S mutation may have reduced neuroprotective capacity and may contribute to the development of PD pathology.
Collapse
Affiliation(s)
| | - Frank Wessely
- UK Dementia Research Institute, Cardiff University, Maindy Road, Cardiff CF24 4HQ, UK
| | | | - Federica Rinaldi
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Jane Vowles
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Cathy Browne
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Samuel G Evetts
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Michele T Hu
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sally A Cowley
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK; James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Caleb Webber
- Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK; UK Dementia Research Institute, Cardiff University, Maindy Road, Cardiff CF24 4HQ, UK.
| | | |
Collapse
|
46
|
Zeuner KE, Schäffer E, Hopfner F, Brüggemann N, Berg D. Progress of Pharmacological Approaches in Parkinson's Disease. Clin Pharmacol Ther 2019; 105:1106-1120. [PMID: 30661251 DOI: 10.1002/cpt.1374] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/22/2018] [Indexed: 12/20/2022]
Abstract
The progressive neurodegenerative process in Parkinson's disease (PD) is not restricted to dopaminergic midbrain neurons but involves the entire nervous system. In this review, we outline established treatment options at different disease stages and address new therapeutic approaches. These include, based on recent advances in the understanding of the pathophysiology of PD, genetic and disease-modifying approaches to reduce abnormal accumulation and aggregation of alpha-synuclein (aSYN), mitochondrial dysfunction, and dysfunction of lysosomal proteins. Moreover, we highlight clinical trials to reduce neuroinflammation and increase neurorestoration.
Collapse
Affiliation(s)
- Kirsten E Zeuner
- Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Eva Schäffer
- Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Franziska Hopfner
- Department of Psychiatry and Psychotherapy, Hospital of the University of Munich, Munich, Germany
| | - Norbert Brüggemann
- Department of Neurology, University of Lübeck, Lübeck, Germany.,Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Daniela Berg
- Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
47
|
Huang C, Wu J, Chen D, Jin J, Wu Y, Chen Z. Effects of sulforaphane in the central nervous system. Eur J Pharmacol 2019; 853:153-168. [PMID: 30858063 DOI: 10.1016/j.ejphar.2019.03.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 12/12/2022]
Abstract
Sulforaphane (SFN) is an active component extracted from vegetables like cauliflower and broccoli. Activation of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling is a common mechanism for the anti-oxidative and anti-inflammatory activity of some herb-derived compounds, such as icariin and berberine. However, due to its peculiar ability in Nrf2 activation, SFN is recognized as an activator of Nrf2 and recommended as a supplementation for prevention and/or treatment of disorders like neoplasm and heart failure. In the central nervous system (CNS), the prophylactic and/or therapeutic effects of SFN have been revealed in recent years. For example, it has been reported to prevent the progression of Alzheimer's disease, Parkinson's disease, cerebral ischemia, Huntington's disease, multiple sclerosis, epilepsy, and psychiatric disorders via promotion of neurogenesis or inhibition of oxidative stress and neuroinflammation. SFN is also implicated in reversing cognition, learning, and memory impairment in rodents induced by scopolamine, lipopolysaccharide, okadaic acid, and diabetes. In models of neurotoxicity, SFN has been shown to suppress neurotoxicity induced by a wide range of toxic factors, such as hydrogen peroxide, prion protein, hyperammonemia, and methamphetamine. To date, no consolidated source of knowledge about the pharmacological effects of SFN in the CNS has been presented in the literature. In this review, we summarize and discuss the pharmacological effects of SFN as well as their possible mechanisms in prevention and/or therapy of disorders afflicting the CNS, aiming to get a further insight into how SFN affects the pathophysiological process of CNS disorders.
Collapse
Affiliation(s)
- Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Dongjian Chen
- Invasive Technology Department, Nantong First People's Hospital, the Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu, China
| | - Jie Jin
- Invasive Technology Department, Nantong First People's Hospital, the Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu, China
| | - Yue Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, the Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu, China.
| |
Collapse
|
48
|
Marques BL, Carvalho GA, Freitas EMM, Chiareli RA, Barbosa TG, Di Araújo AGP, Nogueira YL, Ribeiro RI, Parreira RC, Vieira MS, Resende RR, Gomez RS, Oliveira-Lima OC, Pinto MCX. The role of neurogenesis in neurorepair after ischemic stroke. Semin Cell Dev Biol 2019; 95:98-110. [PMID: 30550812 DOI: 10.1016/j.semcdb.2018.12.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Abstract
Stroke consists of an abrupt reduction of cerebral blood flow resulting in hypoxia that triggers an excitotoxicity, oxidative stress, and neuroinflammation. After the ischemic process, neural precursor cells present in the subventricular zone of the lateral ventricle and subgranular zone of the dentate gyrus proliferate and migrate towards the lesion, contributing to the brain repair. The neurogenesis is induced by signal transduction pathways, growth factors, attractive factors for neuroblasts, transcription factors, pro and anti-inflammatory mediators and specific neurotransmissions. However, this endogenous neurogenesis occurs slowly and does not allow a complete restoration of brain function. Despite that, understanding the mechanisms of neurogenesis could improve the therapeutic strategies for brain repair. This review presents the current knowledge about brain repair process after stroke and the perspectives regarding the development of promising therapies that aim to improve neurogenesis and its potential to form new neural networks.
Collapse
Affiliation(s)
- Bruno L Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Gustavo A Carvalho
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Elis M M Freitas
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Raphaela A Chiareli
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Thiago G Barbosa
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Armani G P Di Araújo
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Yanley L Nogueira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Raul I Ribeiro
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Ricardo C Parreira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Mariana S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Renato S Gomez
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Onésia C Oliveira-Lima
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Mauro C X Pinto
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil.
| |
Collapse
|
49
|
Bachiller S, Jiménez-Ferrer I, Paulus A, Yang Y, Swanberg M, Deierborg T, Boza-Serrano A. Microglia in Neurological Diseases: A Road Map to Brain-Disease Dependent-Inflammatory Response. Front Cell Neurosci 2018; 12:488. [PMID: 30618635 PMCID: PMC6305407 DOI: 10.3389/fncel.2018.00488] [Citation(s) in RCA: 487] [Impact Index Per Article: 69.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Microglia represent a specialized population of macrophages-like cells in the central nervous system (CNS) considered immune sentinels that are capable of orchestrating a potent inflammatory response. Microglia are also involved in synaptic organization, trophic neuronal support during development, phagocytosis of apoptotic cells in the developing brain, myelin turnover, control of neuronal excitability, phagocytic debris removal as well as brain protection and repair. Microglial response is pathology dependent and affects to immune, metabolic. In this review, we will shed light on microglial activation depending on the disease context and the influence of factors such as aging, environment or cell-to-cell interaction.
Collapse
Affiliation(s)
- Sara Bachiller
- Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Itzia Jiménez-Ferrer
- Translational Neurogenetics Unit, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Agnes Paulus
- Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Yiyi Yang
- Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Maria Swanberg
- Translational Neurogenetics Unit, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | | |
Collapse
|
50
|
Silencing of TGFβ signalling in microglia results in impaired homeostasis. Nat Commun 2018; 9:4011. [PMID: 30275444 PMCID: PMC6167353 DOI: 10.1038/s41467-018-06224-y] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 08/20/2018] [Indexed: 12/31/2022] Open
Abstract
TGFβ1 has been implicated in regulating functional aspects of several distinct immune cell populations including central nervous system (CNS) resident microglia. Activation and priming of microglia have been demonstrated to contribute to the progression of neurodegenerative diseases and, thus, underlie stringent control by endogenous regulatory factors including TGFβ1. Here, we demonstrate that deletion of Tgfbr2 in adult postnatal microglia does neither result in impairment of the microglia-specific gene expression signatures, nor is microglial survival and maintenance affected. Tgfbr2-deficient microglia were characterised by distinct morphological changes and transcriptome analysis using RNAseq revealed that loss of TGFβ signalling results in upregulation of microglia activation and priming markers. Moreover, protein arrays demonstrated increased secretion of CXCL10 and CCL2 accompanied by activation of immune cell signalling as evidenced by increased phosphorylation of TAK1. Together, these data underline the importance of microglial TGFβ signalling to regulate microglia adaptive changes.
Collapse
|