1
|
Zheng Y, Ren Z, Liu Y, Yan J, Chen C, He Y, Shi Y, Cheng F, Wang Q, Li C, Wang X. T cell interactions with microglia in immune-inflammatory processes of ischemic stroke. Neural Regen Res 2025; 20:1277-1292. [PMID: 39075894 PMCID: PMC11624874 DOI: 10.4103/nrr.nrr-d-23-01385] [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: 08/19/2023] [Revised: 01/17/2024] [Accepted: 03/07/2024] [Indexed: 07/31/2024] Open
Abstract
The primary mechanism of secondary injury after cerebral ischemia may be the brain inflammation that emerges after an ischemic stroke, which promotes neuronal death and inhibits nerve tissue regeneration. As the first immune cells to be activated after an ischemic stroke, microglia play an important immunomodulatory role in the progression of the condition. After an ischemic stroke, peripheral blood immune cells (mainly T cells) are recruited to the central nervous system by chemokines secreted by immune cells in the brain, where they interact with central nervous system cells (mainly microglia) to trigger a secondary neuroimmune response. This review summarizes the interactions between T cells and microglia in the immune-inflammatory processes of ischemic stroke. We found that, during ischemic stroke, T cells and microglia demonstrate a more pronounced synergistic effect. Th1, Th17, and M1 microglia can co-secrete pro-inflammatory factors, such as interferon-γ, tumor necrosis factor-α, and interleukin-1β, to promote neuroinflammation and exacerbate brain injury. Th2, Treg, and M2 microglia jointly secrete anti-inflammatory factors, such as interleukin-4, interleukin-10, and transforming growth factor-β, to inhibit the progression of neuroinflammation, as well as growth factors such as brain-derived neurotrophic factor to promote nerve regeneration and repair brain injury. Immune interactions between microglia and T cells influence the direction of the subsequent neuroinflammation, which in turn determines the prognosis of ischemic stroke patients. Clinical trials have been conducted on the ways to modulate the interactions between T cells and microglia toward anti-inflammatory communication using the immunosuppressant fingolimod or overdosing with Treg cells to promote neural tissue repair and reduce the damage caused by ischemic stroke. However, such studies have been relatively infrequent, and clinical experience is still insufficient. In summary, in ischemic stroke, T cell subsets and activated microglia act synergistically to regulate inflammatory progression, mainly by secreting inflammatory factors. In the future, a key research direction for ischemic stroke treatment could be rooted in the enhancement of anti-inflammatory factor secretion by promoting the generation of Th2 and Treg cells, along with the activation of M2-type microglia. These approaches may alleviate neuroinflammation and facilitate the repair of neural tissues.
Collapse
Affiliation(s)
- Yuxiao Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zilin Ren
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Juntang Yan
- Library, Beijing University of Chinese Medicine, Beijing, China
| | - Congai Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanhui He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyu Shi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fafeng Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingguo Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Changxiang Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xueqian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Nakamura R, Matsuda A, Higashi Y, Hayashi Y, Konishi M, Saito M, Akizawa T. An 11-mer Synthetic Peptide Suppressing Aggregation of Aβ25-35 and Resolving Its Aggregated Form Improves Test Performance in an Aβ25-35-Induced Alzheimer's Mouse Model. Biomolecules 2024; 14:1234. [PMID: 39456166 PMCID: PMC11506537 DOI: 10.3390/biom14101234] [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: 08/03/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
There is a high demand for the development of drugs against Alzheimer's disease (AD), which is related to the misfolding and aggregation of Amyloid-β (Aβ), due to the increasing number of patients with AD. In our present study, we aimed to assess the aggregation inhibitory effect of various synthetic YS-peptides on Aβ25-35 to identify an applicable peptide for clinical use for AD treatment and prevention. Suppression and aggregate resolution activities of YS-peptides against Aβ25-35 were evaluated using a Thioflavin T assay and scanning electron microscopy (SEM). Structure-activity relationship studies revealed that YS-RD11 (RETLVYLTHLD) and YS-RE16 (RETLVYLTHLDYDDTE) showed suppression and aggregate-resolution activities. The effect of YS-peptides on phagocytosis in microglial cells (BV-2 cells) demonstrated that YS-RD11 and YS-RE16 activated the phagocytic ability of microglia. In the Aβ25-35-induced AD mouse model, YS-RD11 prevented and improved the deficits in short-term memory. In conclusion, YS-RD11 is a suitable candidate therapeutic drug against AD and uses a strategy similar to that used for antibodies.
Collapse
Affiliation(s)
- Rina Nakamura
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku 783-8505, Kochi, Japan; (Y.H.); (M.S.)
- O-Force Co., Ltd., 3454 Irino Kuroshio-cho, Hata-gun 789-1931, Kochi, Japan;
| | - Akira Matsuda
- Laboratory of Medicinal and Biochemical Analysis, Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1, Hirokoshingai, Kure 737-0112, Hiroshima, Japan;
| | - Youichirou Higashi
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku 783-8505, Kochi, Japan; (Y.H.); (M.S.)
| | - Yoshihiro Hayashi
- O-Force Co., Ltd., 3454 Irino Kuroshio-cho, Hata-gun 789-1931, Kochi, Japan;
- Equipment Support Planning Office, Kochi University, Kohasu, Oko-cho, Nankoku 783-8505, Kochi, Japan
| | - Motomi Konishi
- Department of Integrative Pharmacy, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata 573-0101, Osaka, Japan;
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku 783-8505, Kochi, Japan; (Y.H.); (M.S.)
| | - Toshifumi Akizawa
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku 783-8505, Kochi, Japan; (Y.H.); (M.S.)
- O-Force Co., Ltd., 3454 Irino Kuroshio-cho, Hata-gun 789-1931, Kochi, Japan;
| |
Collapse
|
3
|
Živančević K, Aru B, Demir A, Radenović L, Andjus P, Demirel GY. Zn 0-Induced Cytotoxicity and Mitochondrial Stress in Microglia: Implications of the Protective Role of Immunoglobulin G In Vitro. Balkan Med J 2024; 41:348-356. [PMID: 39129512 PMCID: PMC11588912 DOI: 10.4274/balkanmedj.galenos.2024.2024-4-119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/04/2024] [Indexed: 08/13/2024] Open
Abstract
Background Zinc (Zn), an essential micronutrient, regulates and maintains neurological functions. However, both Zn deficiency and excess can cause oxidative stress and neurodegenerative diseases. As previously reported, immunoglobulin G (IgG) can modulate oxidative stress in various disorders. Aims To investigate whether IgG treatment can alleviate oxidative stress caused by Zn0 on microglia in vitro. Study Design In vitro study. Methods The feasibility of Zn0 treatment was evaluated using the MTS assay. Oxidative stress following treatment with Zn0, either alone or with IgG supplementation, was determined with dihydrorhodamine 123 staining. Flow cytometry was employed to ascertain the intracellular protein levels of TRIM21, PINK, PARKIN, MFN2, Beclin-1, and active LC3B. Methods The feasibility of Zn0 treatment was evaluated using the MTS assay. Oxidative stress following treatment with Zn0, either alone or with IgG supplementation, was determined with dihydrorhodamine 123 staining. Flow cytometry was employed to ascertain the intracellular protein levels of TRIM21, PINK, PARKIN, MFN2, Beclin-1, and active LC3B. Results: In silico screening confirmed the association between Zn0 cytotoxicity and apoptosis. Furthermore, oxidative stress was identified as a critical mechanism that underlies Zn0 neurotoxicity. The in silico analysis revealed that Zn can interact with the constant region of the Ig heavy chain, suggesting a potential role for IgG in alleviating Zn0-induced cytotoxicity. Experimental findings supported this hypothesis, as IgG administration significantly reduced Zn0-induced mitochondrial stress in a dose-dependent manner. The upregulation of PINK1 levels by Zn0 exposure suggests that mitochondrial injury promotes mitophagy. Interestingly, Zn0 decreased TRIM21 levels, which is reversed by IgG administration. Conclusion These findings elucidate the cellular responses to Zn0 and highlight the potential use of intravenous immunoglobulin in mitigating the adverse effects of acute Zn0 exposure.
Collapse
Affiliation(s)
- Katarina Živančević
- Department for Physiology and Biochemistry, Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Belgrade, Serbia
- Department of Toxicology “Akademik Danilo Soldatović”, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Başak Aru
- Department of Immunology, Yeditepe University Faculty of Medicine, İstanbul, Türkiye
| | - Abdullah Demir
- Department of Immunology, Yeditepe University Faculty of Medicine, İstanbul, Türkiye
- Stem Cell Laboratory, Yeditepe University Training and Research Hospital, İstanbul, Türkiye
| | - Lidija Radenović
- Department for Physiology and Biochemistry, Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Pavle Andjus
- Department for Physiology and Biochemistry, Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Gülderen Yanıkkaya Demirel
- Department of Immunology, Yeditepe University Faculty of Medicine, İstanbul, Türkiye
- Stem Cell Laboratory, Yeditepe University Training and Research Hospital, İstanbul, Türkiye
| |
Collapse
|
4
|
Lin B, Wang M, Chen X, Chai L, Ni J, Huang J. Involvement of P2X7R-mediated microglia polarization and neuroinflammation in the response to electroacupuncture on post-stroke memory impairment. Brain Res Bull 2024; 212:110967. [PMID: 38670470 DOI: 10.1016/j.brainresbull.2024.110967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
PURPOSE Post-stroke cognitive impairment (PSCI) is a common complication of ischemic stroke episodes. Memory impairment is an important component of the poststroke cognitive syndrome. Microglial activation plays a critical role in stroke-induced neuroinflammation. Previous studies have reported that electroacupuncture (EA) provides neuroprotective effects by reducing the expression levels of the Purinergic receptor P2X ligand-gated ion channel 7 (P2X7) and inhibiting neuroinflammation in rat model of ischemic stroke. Further understanding of the role and connections between P2X7R and microglial activation in EA-induced anti-inflammatory can reveal novel targets for post-stroke memory impairment treatment. METHODS A Middle cerebral artery occlusion and reperfusion (MCAO/R) model was established. We used 2'(3')-O-(4-benzoyl) benzoyl ATP (BzATP) as a P2X7R agonist. Following MCAO/R injury, the rats underwent EA therapy at the Baihui (DU20) and Shenting (DU24) acupoints for seven consecutive days. The Barnes maze test was used to evaluate memory function. Following intervention, a T2 weighted images (T2WI) scan was performed to identify changes in cerebral infarction volume in MCAO/R rats. The levels of Interleukin-1β (IL-1β), Interleukin-6 (IL-6) and Interleukin-4 (IL-4), Interleukin-10 (IL-10) in the peri-infarct hippocampal were examined by ELISA. Immunofluorescence was employed to evaluate Iba-1+ / P2X7R+, Iba-1+/ iNOS+ and Iba-1+/ Arg-1+ cell populations in the peri-infarct hippocampal DG area. The protein expression of P2X7R, Nuclear factor E2-related factor 2 (Nrf2), Recombinant nlr family, pyrin domain containing protein 3 (NLRP3), Inducible nitric oxide synthase (iNOS) and Arginase-1 (Arg-1) in the peri-infarct hippocampal were investigated using western blot assays. Besides, we also measured the levels of reactive oxygen species (ROS), superoxide dismutase (SOD) and malondialdehyde (MDA). RESULTS We found EA treatment reduced inflammation and oxidative stress, which is consistent with a decrease in P2X7R expression and improved learning and memory functions. In contrast, we found BzATP enhanced inflammation and oxidative stress. Moreover, our results showed EA treatment up-regulated Nrf2, down-regulated NLRP3, and promoted microglia M2 polarization. Finally, EA-mediated positive effects were reversed by intracerebroventricular injection of BzATP, which is consistent with an increase in P2X7R expression. CONCLUSION EA ameliorates memory impairment in a rat model of ischemic stroke by reducing inflammation and ROS through the inhibition of P2X7R expression. In turn, this mechanism regulates Nrf2 and NLRP3 expression, suggesting EA is beneficial for ischemic stroke treatment using P2X7R as target.
Collapse
Affiliation(s)
- Bingbing Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Mengxue Wang
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiaocheng Chen
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Linsong Chai
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jinglei Ni
- Key Laboratory of Orthopedics & Traumatology of Traditional Chinese Medicine and Rehabilitation, Ministry of Education, China
| | - Jia Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.
| |
Collapse
|
5
|
Shippy DC, Oliai SF, Ulland TK. Zinc utilization by microglia in Alzheimer's disease. J Biol Chem 2024; 300:107306. [PMID: 38648940 PMCID: PMC11103939 DOI: 10.1016/j.jbc.2024.107306] [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: 02/18/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia defined by two key pathological characteristics in the brain, amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau. Microglia, the primary innate immune cells of the central nervous system (CNS), provide neuroprotection through Aβ and tau clearance but may also be neurotoxic by promoting neuroinflammation to exacerbate Aβ and tau pathogenesis in AD. Recent studies have demonstrated the importance of microglial utilization of nutrients and trace metals in controlling their activation and effector functions. Trace metals, such as zinc, have essential roles in brain health and immunity, and zinc dyshomeostasis has been implicated in AD pathogenesis. As a result of these advances, the mechanisms by which zinc homeostasis influences microglial-mediated neuroinflammation in AD is a topic of continuing interest since new strategies to treat AD are needed. Here, we review the roles of zinc in AD, including zinc activation of microglia, the associated neuroinflammatory response, and the application of these findings in new therapeutic strategies.
Collapse
Affiliation(s)
- Daniel C Shippy
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Sophia F Oliai
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Tyler K Ulland
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA; Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA.
| |
Collapse
|
6
|
Wang Y, Song Y, Zhang L, Huang X. The paradoxical role of zinc on microglia. J Trace Elem Med Biol 2024; 83:127380. [PMID: 38171037 DOI: 10.1016/j.jtemb.2023.127380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024]
Abstract
Zinc is an essential trace element for humans, and its homeostasis is essential for the health of the central nervous system. Microglia, the resident immune cells in the central nervous system, play the roles of sustaining, nourishing, and immune surveillance. Microglia are sensitive to microenvironment changes and are easily activated to M1 phenotype to enhance disease progression or the M2 phenotype to improve peripheral nerves injury repair. Zinc is requisite for microglial activation, However, the cytotoxicity outcome of zinc against microglia, the activated microglia phenotype, and activated microglia function are ambiguous. Herein, we have reviewed the neurological function of zinc and microglia, particularly the ambiguous role of zinc on microglia. We also pay attention to the role of zinc homeostasis on microglial function within the central nervous system disease. Finally, we observe the relationship between zinc and microglia, attempting to design new therapeutic measures against major nervous system disorders.
Collapse
Affiliation(s)
- Yehong Wang
- Graduate Faculty, Xi'an Physical Education University, Xi'an 710068, PR China; Hunan Provincial Key Laboratory of Dong Medicine, Ethnic Medicine Research Center, Hunan University of Medicine, Huaihua 418000, PR China
| | - Yi Song
- Department of Neurosurgery, Chongqing University Three Gorges Hospital, Chongqing 404100, PR China.
| | - Lingdang Zhang
- Department of Neurosurgery, Chongqing University Three Gorges Hospital, Chongqing 404100, PR China
| | - Xiao Huang
- Hunan Provincial Key Laboratory of Dong Medicine, Ethnic Medicine Research Center, Hunan University of Medicine, Huaihua 418000, PR China.
| |
Collapse
|
7
|
Yang X, Li W, Ding M, Liu KJ, Qi Z, Zhao Y. Contribution of zinc accumulation to ischemic brain injury and its mechanisms about oxidative stress, inflammation, and autophagy: an update. Metallomics 2024; 16:mfae012. [PMID: 38419293 DOI: 10.1093/mtomcs/mfae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024]
Abstract
Ischemic stroke is a leading cause of death and disability worldwide, and presently, there is no effective neuroprotective therapy. Zinc is an essential trace element that plays important physiological roles in the central nervous system. Free zinc concentration is tightly regulated by zinc-related proteins in the brain under normal conditions. Disruption of zinc homeostasis, however, has been found to play an important role in the mechanism of brain injury following ischemic stroke. A large of free zinc releases from storage sites after cerebral ischemia, which affects the functions and survival of nerve cells, including neurons, astrocytes, and microglia, resulting in cell death. Ischemia-triggered intracellular zinc accumulation also disrupts the function of blood-brain barrier via increasing its permeability, impairing endothelial cell function, and altering tight junction levels. Oxidative stress and neuroinflammation have been reported to be as major pathological mechanisms in cerebral ischemia/reperfusion injury. Studies have showed that the accumulation of intracellular free zinc could impair mitochondrial function to result in oxidative stress, and form a positive feedback loop between zinc accumulation and reactive oxygen species production, which leads to a series of harmful reactions. Meanwhile, elevated intracellular zinc leads to neuroinflammation. Recent studies also showed that autophagy is one of the important mechanisms of zinc toxicity after ischemic injury. Interrupting the accumulation of zinc will reduce cerebral ischemia injury and improve neurological outcomes. This review summarizes the role of zinc toxicity in cellular and tissue damage following cerebral ischemia, focusing on the mechanisms about oxidative stress, inflammation, and autophagy.
Collapse
Affiliation(s)
- Xueqi Yang
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
- Beijing Geriatric Medical Research Center, Beijing 100053, China
| | - Wei Li
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
- Beijing Geriatric Medical Research Center, Beijing 100053, China
| | - Mao Ding
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
| | - Ke Jian Liu
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Zhifeng Qi
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
- Beijing Geriatric Medical Research Center, Beijing 100053, China
| | - Yongmei Zhao
- Institute of Cerebrovascular Disease Research, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China
- Beijing Geriatric Medical Research Center, Beijing 100053, China
| |
Collapse
|
8
|
Yang X, Wu J, Cheng H, Chen S, Wang J. DEXMEDETOMIDINE AMELIORATES ACUTE BRAIN INJURY INDUCED BY MYOCARDIAL ISCHEMIA-REPERFUSION VIA UPREGULATING THE HIF-1 PATHWAY. Shock 2023; 60:678-687. [PMID: 37647083 DOI: 10.1097/shk.0000000000002217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
ABSTRACT Objective: Neurological complications after myocardial ischemia/reperfusion (IR) injury remain high and seriously burden patients and their families. Dexmedetomidine (Dex), an α 2 agonist, is endowed with analgesic-sedative and anti-inflammatory effects. Therefore, our study aims to explore the mechanism and effect of Dex on brain damage after myocardial IR injury. Methods C57BL/6 mice were randomly divided into sham, IR, and IR + Dex groups, and myocardial IR models were established. The impact of Dex on brain injury elicited by myocardial IR was assessed via ELISA for inflammatory factors in serum and brain; Evans blue for blood-brain barrier permeability; hematoxylin-eosin staining for pathological injury in brain; immunofluorescence for microglia activation in brain; Morris water maze for cognitive dysfunction; western blot for the expression level of HIF-1α, occludin, cleaved caspase-3, NF-κB p65, and p-NF-κB p65 in the brain. In addition, HIF-1α knockout mice were used to verify whether the neuroprotective function of Dex is associated with the HIF-1 pathway. Results: Dex was capable of reducing myocardial IR-induced brain damage including inflammatory factor secretion, blood-brain barrier disruption, neuronal edema, microglial activation, and acute cognitive dysfunction. However, the protective role of Dex was attenuated in HIF-1α knockout mice. Conclusion: Dex protects against myocardial IR-induced brain injury, and the neuroprotection of Dex is at least partially dependent on the activation of the HIF-1 pathway.
Collapse
Affiliation(s)
- Xue Yang
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | | | | | | | | |
Collapse
|
9
|
Chatila ZK, Yadav A, Mares J, Flowers X, Yun TD, Rashid M, Talcoff R, Pelly Z, Zhang Y, De Jager PL, Teich A, Costa R, Gomez EA, Martins G, Alcalay R, Vonsattel JP, Menon V, Bradshaw EM, Przedborski S. RNA- and ATAC-sequencing Reveals a Unique CD83+ Microglial Population Focally Depleted in Parkinson's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.540842. [PMID: 37292857 PMCID: PMC10245789 DOI: 10.1101/2023.05.17.540842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
All brain areas affected in Parkinson's disease (PD) show an abundance of microglia with an activated morphology together with increased expression of pro-inflammatory cytokines, suggesting that neuroinflammation may contribute to the neurodegenerative process in this common and incurable disorder. We applied a single nucleus RNA- and ATAC-sequencing approach using the 10x Genomics Chromium platform to postmortem PD samples to investigate microglial heterogeneity in PD. We created a multiomic dataset using substantia nigra (SN) tissues from 19 PD donors and 14 non-PD controls (NPCs), as well as three other brain regions from the PD donors which are differentially affected in this disease: the ventral tegmental area (VTA), substantia inominata (SI), and hypothalamus (HypoTs). We identified thirteen microglial subpopulations within these tissues as well as a perivascular macrophage and a monocyte population, of which we characterized the transcriptional and chromatin repertoires. Using this data, we investigated whether these microglial subpopulations have any association with PD and whether they have regional specificity. We uncovered several changes in microglial subpopulations in PD, which appear to parallel the magnitude of neurodegeneration across these four selected brain regions. Specifically, we identified that inflammatory microglia in PD are more prevalent in the SN and differentially express PD-associated markers. Our analysis revealed the depletion of a CD83 and HIF1A- expressing microglial subpopulation, specifically in the SN in PD, that has a unique chromatin signature compared to other microglial subpopulations. Interestingly, this microglial subpopulation has regional specificity to the brainstem in non-disease tissues. Furthermore, it is highly enriched for transcripts of proteins involved in antigen presentation and heat-shock proteins, and its depletion in the PD SN may have implications for neuronal vulnerability in disease.
Collapse
|
10
|
Nakamura R, Konishi M, Higashi Y, Saito M, Akizawa T. Five-mer peptides prevent short-term spatial memory deficits in Aβ25-35-induced Alzheimer's model mouse by suppressing Aβ25-35 aggregation and resolving its aggregate form. Alzheimers Res Ther 2023; 15:83. [PMID: 37076912 PMCID: PMC10114458 DOI: 10.1186/s13195-023-01229-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 04/10/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND The development of drugs for Alzheimer's disease (AD), which is related to the misfolding and aggregation of amyloid-β (Aβ), is high in demand due to the growing number of AD patients. In this study, we screened 22 kinds of 5-mer synthetic peptides derived from the Box A region of Tob1 protein to find a peptide effective against Aβ aggregation. METHODS A Thioflavin T (ThT) assay was performed to evaluate aggregation and screen aggregation inhibitors. Male ICR mice (6 weeks old) were administered saline, 9 nmol Aβ25-35, or a mixture of 9 nmol Aβ25-35 and 9 nmol GSGFK in the right lateral ventricle. Short-term spatial memory was assessed through Y-maze. Microglia cells (BV-)2 cells were plated on 24-well plates (4 × 104 cells/well) and incubated for 48 h, and then, the cells were treated with 0.01, 0.05, 0.1, 0.2, or 0.5 mM GSGFK. After incubation for 24 h, bead uptake was evaluated using a laser confocal microscope and Cytation 5. RESULTS We found two kinds of peptides, GSGNR and GSGFK, that were not only suppressed by aggregation of Aβ25-35 but also resolved the aggregated Aβ25-35. Results obtained from the Y-maze test on an Aβ25-35-induced AD model mouse indicated that GSGFK prevents the deficits in short-term memory induced by Aβ25-35. The effect of GSGFK on phagocytosis in BV-2 cells proved that GSGFK activates the phagocytic ability of microglia. CONCLUSIONS In conclusion, 5-mer peptides prevent short-term memory deficit in Aβ25-35 induced AD model mouse by reducing the aggregated Aβ25-35. They may also upregulate the phagocytic ability of microglia, which makes 5-mer peptides suitable candidates as therapeutic drugs against AD.
Collapse
Affiliation(s)
- Rina Nakamura
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
- O-Force Co., Ltd, 3454 Irino Kuroshio-Cho, Hata-Gun, Kochi, 789-1931, Japan
| | - Motomi Konishi
- Department of Integrative Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka, 573-0101, Japan
| | - Youichirou Higashi
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Toshifumi Akizawa
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan.
- O-Force Co., Ltd, 3454 Irino Kuroshio-Cho, Hata-Gun, Kochi, 789-1931, Japan.
| |
Collapse
|
11
|
Neuroprotective effects of carnosine in a mice stroke model concerning oxidative stress and inflammatory response. J Neurol Sci 2023; 447:120608. [PMID: 36906993 DOI: 10.1016/j.jns.2023.120608] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 02/14/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Carnosine (β-alanyl-L-histidine) is a natural dipeptide with multiple neuroprotective properties. Previous studies have advertised that carnosine scavenges free radicals and displays anti-inflammatory activity. However, the underlying mechanism and the efficacies of its pleiotropic effect on prevention remained obscure. In this study, we aimed to investigate the anti-oxidative, anti-inflammative, and anti-pyroptotic effects of carnosine in the transient middle cerebral artery occlusion (tMCAO) mouse model. After a daily pre-treatment of saline or carnosine (1000 mg / kg / day) for 14 days, mice (n = 24) were subjected to tMCAO for 60 min and continuously treated with saline or carnosine for additional 1 and 5 days after reperfusion. The administration of carnosine significantly decreased infarct volume 5 days after the tMCAO (*p < 0.05) and effectively suppressed the expression of 4-HNE, 8-OHdG, Nitrotyrosine 5 days, and RAGE 5 days after tMCAO. Moreover, the expression of IL-1β was also significantly suppressed 5 days after tMCAO. Our present findings demonstrated that carnosine effectively relieves oxidative stress caused by ischemic stroke and significantly attenuates neuroinflammatory responses related to IL-1β, suggesting that carnosine can be a promising therapeutic strategy for ischemic stroke.
Collapse
|
12
|
Chen LL, Fan YG, Zhao LX, Zhang Q, Wang ZY. The metal ion hypothesis of Alzheimer's disease and the anti-neuroinflammatory effect of metal chelators. Bioorg Chem 2023; 131:106301. [PMID: 36455485 DOI: 10.1016/j.bioorg.2022.106301] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/13/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD), characterized by the β-amyloid protein (Aβ) deposition and tau hyperphosphorylation, is the most common dementia with uncertain etiology. The clinical trials of Aβ monoclonal antibody drugs have almost failed, giving rise to great attention on the other etiologic hypothesis regarding AD such as metal ions dysmetabolism and chronic neuroinflammation. Mounting evidence revealed that the metal ions (iron, copper, and zinc) were dysregulated in the susceptible brain regions of AD patients, which was highly associated with Aβ deposition, tau hyperphosphorylation, neuronal loss, as well as neuroinflammation. Further studies uncovered that iron, copper and zinc could not only enhance the production of Aβ but also directly bind to Aβ and tau to promote their aggregations. In addition, the accumulation of iron and copper could respectively promote ferroptosis and cuproptosis. Therefore, the metal ion chelators were recognized as promising agents for treating AD. This review comprehensively summarized the effects of metal ions on the Aβ dynamics and tau phosphorylation in the progression of AD. Furthermore, taking chronic neuroinflammation contributes to the progression of AD, we also provided a summary of the mechanisms concerning metal ions on neuroinflammation and highlighted the metal ion chelators may be potential agents to alleviate neuroinflammation under the condition of AD. Nevertheless, more investigations regarding metal ions on neuroinflammation should be taken into practice, and the effects of metal ion chelators on neuroinflammation should gain more attention. Running title: Metal chelators against neuroinflammation.
Collapse
Affiliation(s)
- Li-Lin Chen
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Qi Zhang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| |
Collapse
|
13
|
Microglial Activation in Metal Neurotoxicity: Impact in Neurodegenerative Diseases. BIOMED RESEARCH INTERNATIONAL 2023; 2023:7389508. [PMID: 36760476 PMCID: PMC9904912 DOI: 10.1155/2023/7389508] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/04/2023]
Abstract
Neurodegenerative processes encompass a large variety of diseases with different pathological patterns and clinical features, such as Alzheimer's and Parkinson's diseases. Exposure to metals has been hypothesized to increase oxidative stress in brain cells leading to cell death and neurodegeneration. Neurotoxicity of metals has been demonstrated by several in vitro and in vivo experimental studies, and most probably, each metal has its specific pathway to trigger cell death. As a result, exposure to essential metals, such as manganese, iron, copper, zinc, and cobalt, and nonessential metals, including lead, aluminum, and cadmium, perturbs metal homeostasis at the cellular and organism levels leading to neurodegeneration. In this contribution, a comprehensive review of the molecular mechanisms by which metals affect microglia physiology and signaling properties is presented. Furthermore, studies that validate the disruption of microglia activation pathways as an essential mechanism of metal toxicity that can contribute to neurodegenerative disease are also presented and discussed.
Collapse
|
14
|
Guo H, Zhang W, Wang Z, Li Z, Zhou J, Yang Z. Dexmedetomidine post-conditioning protects blood-brain barrier integrity by modulating microglia/macrophage polarization via inhibiting NF-κB signaling pathway in intracerebral hemorrhage. Front Mol Neurosci 2022; 15:977941. [PMID: 36172260 PMCID: PMC9512049 DOI: 10.3389/fnmol.2022.977941] [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: 06/25/2022] [Accepted: 08/10/2022] [Indexed: 11/18/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is one of the most devastating forms of stroke. Dexmedetomidine (DEX) has shown certain neuroprotective roles in ICH. Nevertheless, the details concerning the underlying molecular mechanism of DEX’s protective effects still need further elucidation. Herein, a model of ICH was established. The rats were randomly divided into the sham group, the ICH group, and the ICH + DEX group. Neurological outcomes, neuronal injury, and apoptosis were evaluated. Brain water content, Evans blue extravasation, and the expression of tight junction-associated proteins were also detected to assess the blood-brain barrier (BBB) integrity. Subsequently, the microglia/macrophage polarization state and inflammatory cytokine levels were observed. To further explore the underlying mechanism, NF-κB signaling pathway-associated proteins were detected. The results showed that DEX exerted neuroprotective effects against ICH-induced neurological deficits. DEX significantly increased the numbers of the surviving neurons and ameliorated neuronal cell loss and apoptosis in ICH. The rats that received the DEX displayed a lower level of brain water content and EB extravasation, moreover, ZO-1, occludin, and claudin-5 were markedly increased by DEX. Additionally, DEX facilitated M2 microglia/macrophage polarization, the M1-associated markers were reduced by DEX, while the M2-associated identification significantly increased. We found that DEX dramatically diminished pro-inflammatory cytokines expression, simultaneously promoting anti-inflammatory cytokines expression. DEX inhibited nuclear translocation of NF-κB in ICH rats. Our data suggest that DEX post-conditioning protects BBB integrity by modulating microglia/macrophage polarization via inhibiting the NF-κB signaling pathway in ICH.
Collapse
Affiliation(s)
- Hao Guo
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China.,The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Weiwei Zhang
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China
| | - Zhi Wang
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China
| | - Zhishan Li
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China
| | - Jing Zhou
- Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Zhaoyu Yang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Institute of Integrative Medicine, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Wong KA, Benowitz LI. Retinal Ganglion Cell Survival and Axon Regeneration after Optic Nerve Injury: Role of Inflammation and Other Factors. Int J Mol Sci 2022; 23:ijms231710179. [PMID: 36077577 PMCID: PMC9456227 DOI: 10.3390/ijms231710179] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022] Open
Abstract
The optic nerve, like most pathways in the mature central nervous system, cannot regenerate if injured, and within days, retinal ganglion cells (RGCs), the neurons that extend axons through the optic nerve, begin to die. Thus, there are few clinical options to improve vision after traumatic or ischemic optic nerve injury or in neurodegenerative diseases such as glaucoma, dominant optic neuropathy, or optic pathway gliomas. Research over the past two decades has identified several strategies to enable RGCs to regenerate axons the entire length of the optic nerve, in some cases leading to modest reinnervation of di- and mesencephalic visual relay centers. This review primarily focuses on the role of the innate immune system in improving RGC survival and axon regeneration, and its synergy with manipulations of signal transduction pathways, transcription factors, and cell-extrinsic suppressors of axon growth. Research in this field provides hope that clinically effective strategies to improve vision in patients with currently untreatable losses could become a reality in 5-10 years.
Collapse
Affiliation(s)
- Kimberly A. Wong
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Correspondence: (K.A.W.); (L.I.B.)
| | - Larry I. Benowitz
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: (K.A.W.); (L.I.B.)
| |
Collapse
|
16
|
Morimoto M, Nakano T, Egashira S, Irie K, Matsuyama K, Wada M, Nakamura Y, Shigemori Y, Ishikura H, Yamashita Y, Hayakawa K, Sano K, Mishima K. Haptoglobin Regulates Macrophage/Microglia-Induced Inflammation and Prevents Ischemic Brain Damage Via Binding to HMGB1. J Am Heart Assoc 2022; 11:e024424. [PMID: 35243897 PMCID: PMC9075294 DOI: 10.1161/jaha.121.024424] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background HMGB1 (high-mobility group box 1) is known to worsen the functional prognosis after cerebral ischemia. Hp (haptoglobin) binds and sequesters HMGB1. Furthermore, Hp-HMGB1 complexes are rapidly cleared by scavenger receptors on macrophages/microglia and modulate polarization of macrophages/microglia toward the M2 phenotype. Therefore, Hp may prevent aggravation by HMGB1 after cerebral ischemia and promote tissue repair by M2 macrophages/microglia. The aim of this study was to investigate the effects of Hp on ischemic brain damage induced by a high systemic HMGB1 level in mice subjected to 4 hours of middle cerebral artery occlusion (MCAO). Methods and Results One day after MCAO, Hp was administered intraperitoneally at a dose of 20 or 200 U/kg once daily for 7 days. Neurological scores, motor coordination, and plasma HMGB1 levels were measured 1, 3, and 7 days after MCAO. Expression of M1 and M2 macrophage/microglia markers, such as CD16/32 and CD206, were evaluated by immunostaining 7 days after MCAO. Treatment with Hp for 7 days improved the neurological score, motor coordination, and survival and prevented brain damage after MCAO. The systemic HMGB1 level increased 1 to 7 days after MCAO and was higher at 7 days than at day 1. Hp significantly decreased the systemic HMGB1 level and increased the M2 phenotype when compared with the M1 phenotype after MCAO. Conclusions Hp improved functional outcomes, including survival, motor function, and brain damage by binding to HMGB1 and modulating the polarization of macrophages/microglia. Hp may be an effective option in the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Mayuka Morimoto
- Faculty of Pharmaceutical Sciences Department of Physiology and Pharmacology Fukuoka University Fukuoka Japan.,Department of Pharmacy Almeida Memorial Hospital Oita Japan
| | - Takafumi Nakano
- Faculty of Pharmaceutical Sciences Department of Physiology and Pharmacology Fukuoka University Fukuoka Japan.,Departments of Radiology and Neurology Neuroprotection Research LaboratoryMassachusetts General Hospital and Harvard Medical School Charlestown MA
| | - Saki Egashira
- Faculty of Pharmaceutical Sciences Department of Physiology and Pharmacology Fukuoka University Fukuoka Japan
| | - Keiichi Irie
- Faculty of Pharmaceutical Sciences Department of Physiology and Pharmacology Fukuoka University Fukuoka Japan
| | - Kiyoshi Matsuyama
- Faculty of Engineering Fukuoka Institute of Technology Fukuoka Japan
| | - Momoka Wada
- Faculty of Engineering Fukuoka Institute of Technology Fukuoka Japan
| | - Yoshihiko Nakamura
- Department of Emergency and Critical Care Medicine Fukuoka University Hospital Fukuoka Japan
| | - Yutaka Shigemori
- Department of Sports Medicine Faculty of Sports and Health Science Fukuoka University Fukuoka Japan
| | - Hiroyasu Ishikura
- Department of Emergency and Critical Care Medicine Fukuoka University Hospital Fukuoka Japan
| | - Yuta Yamashita
- Faculty of Pharmaceutical Sciences Department of Physiology and Pharmacology Fukuoka University Fukuoka Japan
| | - Kazuhide Hayakawa
- Departments of Radiology and Neurology Neuroprotection Research LaboratoryMassachusetts General Hospital and Harvard Medical School Charlestown MA
| | - Kazunori Sano
- Faculty of Pharmaceutical Sciences Department of Physiology and Pharmacology Fukuoka University Fukuoka Japan
| | - Kenichi Mishima
- Faculty of Pharmaceutical Sciences Department of Physiology and Pharmacology Fukuoka University Fukuoka Japan
| |
Collapse
|
17
|
Wu P, Zhou G, Wu X, Lv R, Yao J, Wen Q. P2X7 receptor induces microglia polarization to the M1 phenotype in cancer-induced bone pain rat models. Mol Pain 2022; 18:17448069211060962. [PMID: 35057643 PMCID: PMC8961217 DOI: 10.1177/17448069211060962] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background The transition from pro-inflammatory M1 phenotype to anti-inflammatory M2
phenotype presents a novel therapeutic strategy for chronic pain. Objective We investigated the role of microglia polarization in cancer-induced bone
pain (CIBP), as well as the role of the P2X7 receptor in modulating M1 to M2
polarization. Methods Walker-256 breast cancer cells were administered into tibias of female rats
to induce bone cancer–associated cancer. Results During bone cancer development, the P2X7 receptor and M1 microglia markers
were upregulated. In contrast, inhibition of the P2X7 receptor by BBG, a
blood-brain barrier-permeable P2X7R-specific antagonist, alleviated the pain
and promoted microglia polarization toward the M2 phenotype, while
suppressing the M1 phenotype in vivo and in
vitro. Conclusion P2X7 receptor-mediated spinal microglia polarization is involved in
alleviation of CIBP. Therefore, P2X7R is a potential option for CIBP
treatment.
Collapse
Affiliation(s)
- Ping Wu
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guohua Zhou
- Anesthesiology Department, Dalian Medical University, Dalian, China
| | - Xiaoqi Wu
- Anesthesiology Department, Dalian Medical University, Dalian, China
| | - Run Lv
- Department of Anesthesiology, The first hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaqi Yao
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingping Wen
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
18
|
Yin B, Wang X, Huang T, Jia J. Shared Genetics and Causality Between Decaffeinated Coffee Consumption and Neuropsychiatric Diseases: A Large-Scale Genome-Wide Cross-Trait Analysis and Mendelian Randomization Analysis. Front Psychiatry 2022; 13:910432. [PMID: 35898629 PMCID: PMC9309364 DOI: 10.3389/fpsyt.2022.910432] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Coffee or caffeine consumption has been associated with neuropsychiatric disorders, implying a shared etiology. However, whether these associations reflect causality remains largely unknown. To understand the genetic structure of the association between decaffeinated coffee consumption (DCC) and neuropsychiatric traits, we examined the genetic correlation, causality, and shared genetic structure between DCC and neuropsychiatric traits using linkage disequilibrium score regression, bidirectional Mendelian randomization (MR), and genome-wide cross-trait meta-analysis in large GWAS Consortia for coffee consumption (N = 329,671) and 13 neuropsychiatric traits (sample size ranges from 36,052 to 500,199). We found strong positive genetic correlations between DCC and lifetime cannabis use (LCU; Rg = 0.48, P = 8.40 × 10-19), alcohol use disorder identification test (AUDIT) total score (AUDIT_T; Rg = 0.40, P = 4.63 × 10-13), AUDIT_C score (alcohol consumption component of the AUDIT; Rg = 0.40, P = 5.26 × 10-11), AUDIT_P score (dependence and hazardous-use component of the AUDIT; Rg = 0.28, P = 1.36 × 10-05), and strong negative genetic correlations between DCC and neuroticism (Rg = -0.15, P = 7.27 × 10-05), major depressed diseases (MDD; Rg = -0.15, P = 0.0010), and insomnia (Rg= -0.15, P = 0.0007). In the cross-trait meta-analysis, we identified 6, 5, 1, 1, 2, 31, and 27 shared loci between DCC and Insomnia, LCU, AUDIT_T, AUDIT_C, AUDIT_P, neuroticism, and MDD, respectively, which were mainly enriched in bone marrow, lymph node, cervix, uterine, lung, and thyroid gland tissues, T cell receptor signaling pathway, antigen receptor-mediated signaling pathway, and epigenetic pathways. A large of TWAS-significant associations were identified in tissues that are part of the nervous system, digestive system, and exo-/endocrine system. Our findings further indicated a causal influence of liability to DCC on LCU and low risk of MDD (odds ratio: 0.90, P = 9.06 × 10-5 and 1.27, P = 7.63 × 10-4 respectively). We also observed that AUDIT_T and AUDIT_C were causally related to DCC (odds ratio: 1.83 per 1-SD increase in AUDIT_T, P = 1.67 × 10-05, 1.80 per 1-SD increase in AUDIT_C, P = 5.09 × 10-04). Meanwhile, insomnia and MDD had a causal negative influence on DCC (OR: 0.91, 95% CI: 0.86-0.95, P = 1.51 × 10-04 for Insomnia; OR: 0.93, 95% CI: 0.89-0.99, P = 6.02 × 10-04 for MDD). These findings provided evidence for the shared genetic basis and causality between DCC and neuropsychiatric diseases, and advance our understanding of the shared genetic mechanisms underlying their associations, as well as assisting with making recommendations for clinical works or health education.
Collapse
Affiliation(s)
- Bian Yin
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xinpei Wang
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China.,Center for Intelligent Public Health, Academy for Artificial Intelligence, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China.,Center for Statistical Science, Peking University, Beijing, China
| |
Collapse
|
19
|
Huiliang Z, Mengzhe Y, Xiaochuan W, Hui W, Min D, Mengqi W, Jianzhi W, Zhongshan C, Caixia P, Rong L. Zinc induces reactive astrogliosis through ERK-dependent activation of Stat3 and promotes synaptic degeneration. J Neurochem 2021; 159:1016-1027. [PMID: 34699606 DOI: 10.1111/jnc.15531] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/16/2021] [Accepted: 10/19/2021] [Indexed: 01/06/2023]
Abstract
Reactive astrogliosis is an early event in Alzheimer's disease (AD) brain and plays a key role in synaptic degeneration in AD development. Zinc accumulates in extracellular fraction and synaptosomes in AD human brains with its effect on reactive astrocytes remaining unknown. Through Western blotting, Quantitative polymerase chain reaction (qPCR), and immunofluorescence detection on primary astrocytes treated by zinc and/or zinc chelator, we revealed that zinc induced harmful A1-type reactive astrogliosis in cultured primary astrocytes; the latter, promoted synaptic degeneration in primary neurons. The mechanism investigation showed that zinc induced activation of extracellular regulated protein kinase (ERK) and Janus kinase 2 (JAK2), which phosphorylated signal transduction and transcription activator 3 (Stat3) at serine 727 (S727-Stat3) and tyrosine 705 (Y705-Stat3), respectively, resulting in activation of Stat3. Stat3 phosphorylation at S727 by ERK plays a key role in zinc-induced astrogliosis. These data imply a new molecular mechanism of reactive astrogliosis in AD, in which excessive zinc activates Stat3 through up-regulating ERK signaling pathway.
Collapse
Affiliation(s)
- Zhang Huiliang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Mengzhe
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wang Xiaochuan
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Hui
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Taikang Tongji Hospital, Wuhan, China
| | - Du Min
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wang Mengqi
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wang Jianzhi
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Peng Caixia
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Central Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Rong
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Saito M, Shimizu S, Higashi Y, Shimizu T. [Pharmacological training with small animals in Kochi University]. Nihon Yakurigaku Zasshi 2021; 156:259-264. [PMID: 34470927 DOI: 10.1254/fpj.21004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The use of animals for scientific purposes is still a subject of debate. The current regulatory position in most western countries allows regulated animal use to occur because of the perceived benefits in generating new knowledge. The use of animals for scientific purposes engenders a wide range of ethical perspectives, with some people looking for the complete termination of animal use, and others strongly support their continued use. Although regulatory systems vary from country to country, in most jurisdictions, research and teaching institutions are required to ensure that staff and students using animals for scientific purposes are appropriately trained, that animals are well cared for, and that the ethical review process for projects is robust. In the curriculum of the Kochi Medical School, it is mandatory for all medical students to perform laboratory exercises in the class of Pharmacology. For the purpose of this experimental class it is common to use small animals in these exercises. However, in recent years in many countries, alternative methods to replace the use of small animals have been introduced. Such methods are experiment simulations with the use of computers and they have been used in some medical schools. In this manuscript, I will make an introduction on how we perform pharmacological laboratory exercises with the use of small animals in Kochi Medical School. Additionally, I would like to discuss the necessity of the use of small animals in exercises as part of the training of medical students.
Collapse
Affiliation(s)
- Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University
| | - Shogo Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University
| | | | - Takahiro Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University
| |
Collapse
|
21
|
Dong R, Huang R, Wang J, Liu H, Xu Z. Effects of Microglial Activation and Polarization on Brain Injury After Stroke. Front Neurol 2021; 12:620948. [PMID: 34276530 PMCID: PMC8280287 DOI: 10.3389/fneur.2021.620948] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
Stroke is one of the most common causes of death worldwide. The subsequent development of neuroinflammation and brain edema dramatically increases the risks associated with stroke, leading to a substantial increase in mortality. Although considerable progress has been made in improving cerebral perfusion in the acute phase of stroke, effective treatment options for the subacute and chronic phases associated with cerebral infarction are limited. Microglia, the innate immune cells of the central nervous system (CNS), can be activated and polarized to take on different phenotypes in response to stimulations associated with stroke, including pro-inflammatory and anti-inflammatory phenotypes, which affect the prognosis of stroke. Therefore, investigation of the activation and polarizing mechanisms of microglia plays a critical role in treating stroke. The aim of this article was to investigate the significance of microglial phenotype regulation in stroke treatment by summarizing the activation, polarizing mechanisms, and general microglia characteristics.
Collapse
Affiliation(s)
- Rui Dong
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Renxuan Huang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiaoqi Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hongyu Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongxin Xu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
22
|
Sarkar S. Mechanism of Gene-Environment Interactions Driving Glial Activation in Parkinson's Diseases. Curr Environ Health Rep 2021; 8:203-211. [PMID: 34043217 DOI: 10.1007/s40572-021-00320-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW Parkinson's disease (PD) is the most prevalent motor disorder and is characterized by loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the brain. Though the pathology of PD is well established, the cause of this neuronal loss is not well understood. Approximately 90% of PD cases are sporadic, and the environment plays a significant role in disease pathogenesis. The etiology of PD is highly complex, with neuroinflammation being one of the most critical factors implicated in PD. However, the signaling mechanisms underlying neuroinflammation and its interaction with environmental factors are unclear. RECENT FINDINGS Astroglia and microglia are the two principal cells that play an essential role in maintaining neuronal health in many ways, including through immunological means. Exposure to environmental stressors from various sources affects these glial cells leading to chronic and sustained inflammation. Recent epidemiological studies have identified an interaction among environmental factors and glial genes in Parkinson's disease. Mechanistic studies have shown that exposure to pesticides like rotenone and paraquat, neurotoxic metals like manganese and lead, and even diesel exhaust fumes induce glial activation by regulating various key inflammatory pathways, including the inflammasomes, NOX pathways, and others. This review aims to discuss the recent advances in understanding the mechanism of glial induction in response to environmental stressors and discuss the potential role of gene-environment interaction in driving glial activation.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| |
Collapse
|
23
|
The Role of NADPH Oxidase in Neuronal Death and Neurogenesis after Acute Neurological Disorders. Antioxidants (Basel) 2021; 10:antiox10050739. [PMID: 34067012 PMCID: PMC8151966 DOI: 10.3390/antiox10050739] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 01/22/2023] Open
Abstract
Oxidative stress is a well-known common pathological process involved in mediating acute neurological injuries, such as stroke, traumatic brain injury, epilepsy, and hypoglycemia-related neuronal injury. However, effective therapeutic measures aimed at scavenging free reactive oxygen species have shown little success in clinical trials. Recent studies have revealed that NADPH oxidase, a membrane-bound enzyme complex that catalyzes the production of a superoxide free radical, is one of the major sources of cellular reactive oxygen species in acute neurological disorders. Furthermore, several studies, including our previous ones, have shown that the inhibition of NADPH oxidase can reduce subsequent neuronal injury in neurological disease. Moreover, maintaining appropriate levels of NADPH oxidase has also been shown to be associated with proper neurogenesis after neuronal injury. This review aims to present a comprehensive overview of the role of NADPH oxidase in neuronal death and neurogenesis in multiple acute neurological disorders and to explore potential pharmacological strategies targeting the NADPH-related oxidative stress pathways.
Collapse
|
24
|
Longitudinal PET Imaging of α7 Nicotinic Acetylcholine Receptors with [ 18F]ASEM in a Rat Model of Parkinson's Disease. Mol Imaging Biol 2021; 22:348-357. [PMID: 31286348 DOI: 10.1007/s11307-019-01400-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE The nicotinic acetylcholine alpha-7 receptors (α7R) are involved in a number of neuropsychiatric and neurodegenerative brain disorders such as Parkinson's disease (PD). However, their specific pathophysiologic roles are still unclear. In this context, we studied the evolution of these receptors in vivo by positron emission tomography (PET) imaging using the recently developed tracer 3-(1,4-diazabicyclo[3.2.2]nonan-4-yl)-6-[18F]fluorodibenzo[b,d]thiophene-5,5-dioxide) in a rat model mimicking early stages of PD. PROCEDURES PET imaging of α7R was performed at 3, 7, and 14 days following a partial striatal unilateral lesion with 6-hydroxydopamine in adult rats. After the last imaging experiments, the status of nigro-striatal dopamine neurons as well as different markers of neuroinflammation was evaluated on brain sections by autoradiographic and immunofluorescent experiments. RESULTS We showed an early and transitory rise in α7R expression in the lesioned striatum and substantia nigra, followed by over-expression of several gliosis activation markers in these regions of interest. CONCLUSIONS These findings support a longitudinally follow-up of α7R in animal models of PD and highlight the requirement to use a potential neuroprotective approach through α7R ligands at the early stages of PD.
Collapse
|
25
|
The role of diurnal fluctuations in excitatory amino acid carrier 1 levels in post-ischemic hippocampal Zn 2+ accumulation. Exp Neurol 2020; 336:113538. [PMID: 33253705 DOI: 10.1016/j.expneurol.2020.113538] [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: 08/18/2020] [Revised: 11/04/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Accumulating evidence indicates time-of-day variations in ischemic neuronal injury. Under ischemic conditions, Zn2+ is massively released from hippocampal glutamatergic neurons, and intracellular Zn2+ accumulation results in neuron death. Notably, excitatory amino acid carrier 1 (EAAC1), known as a cysteine transporter, is involved in Zn2+ homeostasis, and its expressions exhibit a diurnal fluctuation. This study aimed to investigate whether time of day of an ischemic insult affects Zn2+ accumulation and neuronal injury and determine whether altered Zn2+ accumulation is modulated by EAAC1 diurnal fluctuation in the hippocampus in a mouse model of ischemic stroke. Mice subjected to transient global ischemia for 40 min at Zeitgeber time 18 (ZT18) (23:00) exhibited reduced Zn2+ accumulation and neuronal death in the hilar region of the hippocampus compared to those at ZT4 (09:00). The EAAC1 protein expression in the hippocampus was increased at ZT18 relative to ZT4. Intracerebroventricular injection of a non-selective excitatory amino acid transporter inhibitor, DL-threo-β-benzyloxyaspartate, or a selective EAAC1 inhibitor, L-aspartic acid β-hydroxamate, increased ischemia-induced Zn2+ accumulation and neuronal death in the hilus at ZT18. These findings suggest that ischemia-induced Zn2+ accumulation displays circadian fluctuations through diurnal variations in EAAC1 expressions and affects susceptibility to ischemic neuronal injury in the hippocampal hilar region.
Collapse
|
26
|
Transient Receptor Potential Melastatin 2 (TRPM2) Inhibition by Antioxidant, N-Acetyl-l-Cysteine, Reduces Global Cerebral Ischemia-Induced Neuronal Death. Int J Mol Sci 2020; 21:ijms21176026. [PMID: 32825703 PMCID: PMC7504640 DOI: 10.3390/ijms21176026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
A variety of pathogenic mechanisms, such as cytoplasmic calcium/zinc influx, reactive oxygen species production, and ionic imbalance, have been suggested to play a role in cerebral ischemia induced neurodegeneration. During the ischemic state that occurs after stroke or heart attack, it is observed that vesicular zinc can be released into the synaptic cleft, and then translocated into the cytoplasm via various cation channels. Transient receptor potential melastatin 2 (TRPM2) is highly distributed in the central nervous system and has high sensitivity to oxidative damage. Several previous studies have shown that TRPM2 channel activation contributes to neuroinflammation and neurodegeneration cascades. Therefore, we examined whether anti-oxidant treatment, such as with N-acetyl-l-cysteine (NAC), provides neuroprotection via regulation of TRPM2, following global cerebral ischemia (GCI). Experimental animals were then immediately injected with NAC (150 mg/kg/day) for 3 and 7 days, before sacrifice. We demonstrated that NAC administration reduced activation of GCI-induced neuronal death cascades, such as lipid peroxidation, microglia and astroglia activation, free zinc accumulation, and TRPM2 over-activation. Therefore, modulation of the TRPM2 channel can be a potential therapeutic target to prevent ischemia-induced neuronal death.
Collapse
|
27
|
Hamada T, Aratake T, Higashi Y, Ueba Y, Shimizu T, Shimizu S, Yawata T, Ueba T, Nakamura R, Akizawa T, Fujieda M, Saito M. Zinc-aggravated M1 microglia regulate astrocytic engulfment via P2×7 receptors. J Trace Elem Med Biol 2020; 61:126518. [PMID: 32353820 DOI: 10.1016/j.jtemb.2020.126518] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 03/03/2020] [Accepted: 03/27/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND Glial cells such as astrocytes and microglia play an important role in the central nervous system via communication between these glial cells. Activated microglia can exhibit either the inflammatory M1 phenotype or the anti-inflammatory M2 phenotype, which influences astrocytic neuroprotective functions, including engulfment of cell debris. Recently, extracellular zinc has been shown to promote the inflammatory M1 phenotype in microglia through intracellular zinc accumulation and reactive oxygen species (ROS) generation. PURPOSE Here, we investigated whether the zinc-enhanced inflammatory M1 phenotype of microglia affects the astrocytic engulfing activity. METHODS Engulfing activity was assessed in astrocytes treated with microglial-conditioned medium (MCM) from lipopolysaccharide (LPS)-activated or from ZnCl2-pretreated LPS-activated M1 microglia. The effect of zinc on microglia phenotype was also validated using the zinc chelator N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN) and the ROS scavenger Trolox. RESULTS Although treatment of astrocytes with LPS showed no significant effect on the engulfing activity, MCM from LPS-induced M1 microglia increased the beads uptake by astrocytes. This increased uptake activity was suppressed when MCM from LPS-induced M1 microglia pretreated with ZnCl2 was applied to astrocytes, which was further abolished by the intracellular zinc chelator TPEN and the ROS scavenger Trolox. In addition, expression of P2×7 receptors (P2×7R) was increased in astrocytes treated with MCM derived from M1 microglia but not in the M1 microglia pretreated with ZnCl2. CONCLUSION These findings suggest that zinc pre-treatment abolishes the ability of LPS-induced M1 microglia to increase the engulfing activity in astrocytes via alteration of astrocytic P2×7R.
Collapse
Affiliation(s)
- Tomoya Hamada
- Department of Pediatrics, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan; Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Takaaki Aratake
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan; Research Fellow of Japan Society for the Promotion of Science, Japan
| | - Youichirou Higashi
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Yusuke Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Takahiro Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Shogo Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Toshio Yawata
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Tetsuya Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Rina Nakamura
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Toshifumi Akizawa
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Mikiya Fujieda
- Department of Pediatrics, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| |
Collapse
|
28
|
Gui X, Wang H, Wu L, Tian S, Wang X, Zheng H, Wu W. Botulinum toxin type A promotes microglial M2 polarization and suppresses chronic constriction injury-induced neuropathic pain through the P2X7 receptor. Cell Biosci 2020; 10:45. [PMID: 32211150 PMCID: PMC7092425 DOI: 10.1186/s13578-020-00405-3] [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: 11/14/2019] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
Background Switching microglial polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype represents a novel therapeutic strategy for neuropathic pain (NP). This study aims to investigate whether botulinum toxin type A (BTX-A) regulates microglial M1/M2 polarization by inhibiting P2X7 expression in a rat model of NP. Results The BTX-A administration elevated pain threshold, induced microglial polarization toward the M2 phenotype, and decreased P2X7 protein level in a rat model of NP induced by chronic compression injury (CCI). Lipopolysaccharide (LPS) was used to activate HAPI rat microglial cells as an in vitro inflammatory model and we demonstrated that BTX-A promoted microglial M2 polarization in LPS-stimulated HAPI microglial cells through suppressing P2X7. Conclusions Our results indicate that BTX-A promotes microglial M2 polarization and suppresses CCI-induced NP through inhibiting P2X7 receptor. These findings provide new insights into the mechanism of BTX-A in relieving NP.
Collapse
Affiliation(s)
- Xianwei Gui
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006 Jiangxi China
| | - Hansen Wang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006 Jiangxi China
| | - Lanxiang Wu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006 Jiangxi China
| | - Sheng Tian
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006 Jiangxi China
| | - Xuan Wang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006 Jiangxi China
| | - Heqing Zheng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006 Jiangxi China
| | - Wei Wu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006 Jiangxi China
| |
Collapse
|
29
|
Zhang D, Lu Z, Zhang Z, Man J, Guo R, Liu C, Wang J. A likely protective effect of dimethyl itaconate on cerebral ischemia/reperfusion injury. Int Immunopharmacol 2019; 77:105924. [PMID: 31678864 DOI: 10.1016/j.intimp.2019.105924] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/26/2019] [Accepted: 09/19/2019] [Indexed: 12/25/2022]
Abstract
As a membrane-permeable derivative of itaconate, dimethyl itaconate (DMI) was recently showed to limit inflammatory response of activated macrophages, and to decrease the generation of reactive oxygen species and reduce cardiac ischemia/reperfusion injury. However, the effect of DMI in the context of cerebral ischemia/reperfusion injury remains unclear. Here, we treated the transient middle cerebral artery occlusion (tMCAO) mice with DMI or saline at the beginning of occlusion, and allowed them to recover for 3 days. We found that DMI obviously decreased the neurologic deficit score. Further, DMI significantly inhibited the toxic conversion of the peri-infarct microglia, and decreased the protein level of interleukin 1β. The present findings suggest that DMI might be recognized as a promising candidate for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Di Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhengfang Lu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhen Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jiang Man
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Ruiming Guo
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Chang Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jianping Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
30
|
Higashi Y, Aratake T, Shimizu S, Shimizu T, Saito M. [Brain zinc dyshomeostasis and glial cells in ischemic stroke]. Nihon Yakurigaku Zasshi 2019; 154:138-142. [PMID: 31527364 DOI: 10.1254/fpj.154.138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Zinc, an essential trace element, plays an important role in a large number of biological functions. In mammalian brain, whereas the majority of brain zinc is bound to proteins including metallothionein, about 5-15% is stored in presynaptic vesicles of glutamatergic neurons throughout the forebrain, especially in the hippocampus, in a relatively free state. Thus, free zinc (Zn2+) concentration in the brain is considered to be regulated in order to maintain normal brain functions such as learning and memory. On the other hand, brain Zn2+ dyshomeostasis has been recognized as a mechanism for neuronal injury in brain disorders including Alzheimer's disease and brain ischemia. In particular, after transient brain ischemia, Zn2+ accumulates in hippocampal neurons via a zinc transport system, or via release from cytosolic zinc-binding proteins, which results in neuronal cell death. Recently, it has been demonstrated that Zn2+ dyshomeostasis also occurs in glial cells such as microglia, astrocytes and oligodendrocytes after brain ischemia. In oligodendrocytes, ischemic insult triggers intracellular Zn2+ accumulation, resulting in cell death via mitochondrial dysfunction. Increased extracellular Zn2+ inhibits astrocytic glutamate uptake. In addition, extracellular Zn2+ massively released from ischemic neurons primes microglia to enhance production of pro-inflammatory cytokines in response to stimuli that trigger M1 activation. This review aims to describe the impact of brain Zn2+ dyshomeostasis on alterations in glial cell survival and functions in post-ischemic brains.
Collapse
Affiliation(s)
| | - Takaaki Aratake
- Department of Pharmacology, Kochi Medical School, Kochi University
| | - Shogo Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University
| | - Takahiro Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University
| |
Collapse
|
31
|
Evangelou E, Gao H, Chu C, Ntritsos G, Blakeley P, Butts AR, Pazoki R, Suzuki H, Koskeridis F, Yiorkas AM, Karaman I, Elliott J, Luo Q, Aeschbacher S, Bartz TM, Baumeister SE, Braund PS, Brown MR, Brody JA, Clarke TK, Dimou N, Faul JD, Homuth G, Jackson AU, Kentistou KA, Joshi PK, Lemaitre RN, Lind PA, Lyytikäinen LP, Mangino M, Milaneschi Y, Nelson CP, Nolte IM, Perälä MM, Polasek O, Porteous D, Ratliff SM, Smith JA, Stančáková A, Teumer A, Tuominen S, Thériault S, Vangipurapu J, Whitfield JB, Wood A, Yao J, Yu B, Zhao W, Arking DE, Auvinen J, Liu C, Männikkö M, Risch L, Rotter JI, Snieder H, Veijola J, Blakemore AI, Boehnke M, Campbell H, Conen D, Eriksson JG, Grabe HJ, Guo X, van der Harst P, Hartman CA, Hayward C, Heath AC, Jarvelin MR, Kähönen M, Kardia SLR, Kühne M, Kuusisto J, Laakso M, Lahti J, Lehtimäki T, McIntosh AM, Mohlke KL, Morrison AC, Martin NG, Oldehinkel AJ, Penninx BWJH, Psaty BM, Raitakari OT, Rudan I, Samani NJ, Scott LJ, Spector TD, Verweij N, Weir DR, Wilson JF, Levy D, Tzoulaki I, Bell JD, Matthews PM, Rothenfluh A, Desrivières S, Schumann G, Elliott P. New alcohol-related genes suggest shared genetic mechanisms with neuropsychiatric disorders. Nat Hum Behav 2019; 3:950-961. [PMID: 31358974 PMCID: PMC7711277 DOI: 10.1038/s41562-019-0653-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 06/11/2019] [Indexed: 12/19/2022]
Abstract
Excessive alcohol consumption is one of the main causes of death and disability worldwide. Alcohol consumption is a heritable complex trait. Here we conducted a meta-analysis of genome-wide association studies of alcohol consumption (g d-1) from the UK Biobank, the Alcohol Genome-Wide Consortium and the Cohorts for Heart and Aging Research in Genomic Epidemiology Plus consortia, collecting data from 480,842 people of European descent to decipher the genetic architecture of alcohol intake. We identified 46 new common loci and investigated their potential functional importance using magnetic resonance imaging data and gene expression studies. We identify genetic pathways associated with alcohol consumption and suggest genetic mechanisms that are shared with neuropsychiatric disorders such as schizophrenia.
Collapse
Affiliation(s)
- Evangelos Evangelou
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - He Gao
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Congying Chu
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Georgios Ntritsos
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Paul Blakeley
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- NIHR Imperial Biomedical Research Centre, ITMAT Data Science Group, Imperial College London, London, UK
| | - Andrew R Butts
- Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Raha Pazoki
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Hideaki Suzuki
- Centre for Restorative Neurosciences, Division of Brain Sciences, Department of Medicine, Hammersmith Campus, Imperial College London, London, UK
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Fotios Koskeridis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Andrianos M Yiorkas
- Department of Life Sciences, Brunel University London, London, UK
- Section of Investigative Medicine, Imperial College London, London, UK
| | - Ibrahim Karaman
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Joshua Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Qiang Luo
- Institute of Science and Technology for Brain-Inspired Intelligence, MOE-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Department of Psychology and the Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | | | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Sebastian E Baumeister
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Chair of Epidemiology, Ludwig-Maximilians-Universitat Munchen, UNIKA-T Augsburg, Augsburg, Germany
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Toni-Kim Clarke
- Department of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Niki Dimou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Katherine A Kentistou
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Penelope A Lind
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and LHealth Technology, Tampere University, Tampere, Finland
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, Finland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas Foundation Trust, London, UK
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Ilja M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mia-Maria Perälä
- Folkhälsan Research Center, Helsinki, Finland
- Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - David Porteous
- Generation Scotland, Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - Scott M Ratliff
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Samuli Tuominen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sébastien Thériault
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, Quebec, Canada
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - John B Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Alexis Wood
- Department of Pediatrics/Nutrition, Baylor College of Medicine, Houston, TX, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juha Auvinen
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Oulunkaari Health Center, Ii, Finland
| | - Chunyu Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Minna Männikkö
- Northern Finland Birth Cohorts, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Lorenz Risch
- Institute of Clinical Chemistry, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Labormedizinisches Zentrum Dr. Risch, Vaduz, Liechtenstein
- Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Harold Snieder
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Juha Veijola
- Department of Psychiatry, Research Unit of Clinical Neuroscience, University of Oulu, Oulu, Finland
- Department of Psychiatry, University Hospital of Oulu, Oulu, Finland
- Medical research Center Oulu, University and University Hospital of Oulu, Oulu, Finland
| | - Alexandra I Blakemore
- Department of Life Sciences, Brunel University London, London, UK
- Section of Investigative Medicine, Imperial College London, London, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- Unit of General Practice, Helsinki University Central Hospital, Helsinki, Finland
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, Greifswald, Germany
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands
| | - Catharina A Hartman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Andrew C Heath
- Department of Psychiatry, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Michael Kühne
- Cardiology Division, University Hospital Basel, Basel, Switzerland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and LHealth Technology, Tampere University, Tampere, Finland
| | - Andrew M McIntosh
- Department of Psychiatry, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Edinburgh, UK
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Albertine J Oldehinkel
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - James F Wilson
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Jimmy D Bell
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, UK
| | - Paul M Matthews
- Centre for Restorative Neurosciences, Division of Brain Sciences, Department of Medicine, Hammersmith Campus, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Adrian Rothenfluh
- Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Departments of Psychiatry, Neurobiology & Anatomy, Human Genetics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Sylvane Desrivières
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Gunter Schumann
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- PONS Research Group, Dept of Psychiatry and Psychotherapy, Campus Charite Mitte, Humboldt University, Berlin, Germany and Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, P.R. China.
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK.
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK.
- UK Dementia Research Institute, Imperial College London, London, UK.
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College Healthcare NHS Trust and Imperial College London, London, UK.
- Health Data Research UK London Substantive Site, London, UK.
| |
Collapse
|
32
|
de Andrade DF, Vukosavljevic B, Hoppe JB, Pohlmann AR, Guterres SS, Windbergs M, Külkamp-Guerreiro I, Salbego CG, Beck RCR. Redispersible Spray-Dried Powder Containing Nanoencapsulated Curcumin: the Drying Process Does Not Affect Neuroprotection In vitro. AAPS PharmSciTech 2019; 20:283. [PMID: 31407115 DOI: 10.1208/s12249-019-1501-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/03/2019] [Indexed: 12/12/2022] Open
Abstract
A redispersible spray-dried formulation containing curcumin-loaded, lipid-core nanocapsules (LNC-C) was developed for oral administration. The neuroprotective activity of curcumin after the spray-drying process was evaluated in vitro. The spray-dried powder (SD-LNC-C) was produced using a drying adjuvant composed of a blend of maltodextrin and L-leucine (90:10 w/w). Acceptable process yield (~ 70%) and drug content (6.5 ± 0.2 mg g-1) were obtained. SD-LNC-C was formed by smooth, spherical-shaped particles, and confocal Raman analysis indicated the distribution of the LNC-C on the surface of the leucine/maltodextrin agglomerates. The surface of the agglomerates was formed by a combination of LNC-C and adjuvants, and laser diffraction showed that SD-LNC-C had adequate aqueous redispersion, with no loss of controlled drug release behaviour of LNC-C. The in vitro curcumin activity against the lipopolysaccharide (LPS)-induced proinflammatory response in organotypic hippocampal slice cultures was evaluated. Both formulations (LNC-C and SD-LNC-C) reduced TNF-α to similar levels. Therefore, neuroprotection of curcumin in vitro may be improved by nanoencapsulation followed by spray-drying, with no loss of this superior performance. Hence, the redispersible spray-dried powder proposed here represents a suitable approach for the development of innovative nanomedicines containing curcumin for the prevention/treatment of neurodegenerative diseases.
Collapse
|
33
|
Furuta T, Ohishi A, Nagasawa K. Intracellular labile zinc is a determinant of vulnerability of cultured astrocytes to oxidative stress. Neurosci Lett 2019; 707:134315. [PMID: 31185281 DOI: 10.1016/j.neulet.2019.134315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 01/08/2023]
Abstract
Recently, we found that treatment of cultured mouse astrocytes of ddY-strain mice (ddY-astrocytes) with 400 μM H2O2 for 24 h increased the intracellular labile zinc level without cell toxicity. In contrast, 170 μM H2O2 for 12 h is reported to kill mouse astrocytes obtained from C57BL/6-strain mice (C57BL/6-astrocytes) with an increase in intracellular labile zinc. To clarify this discrepancy, we compared the intracellular zinc levels and cell toxicity in H2O2-treated ddY- and C57BL/6-astrocytes. Exposure of C57BL/6-astrocytes to 170 or 400 μM H2O2 for 12 h dose-dependently decreased the cell viability and administration of plasma membrane-permeable zinc chelator TPEN prevented the 170 μM H2O2-induced astrocyte death, while neither concentration of H2O2 killed ddY-astrocytes. The intracellular zinc level in H2O2-treated C57BL/6-astrocytes was higher than that in H2O2-treated ddY-astrocytes, and this increase was suppressed by TPEN. There was no apparent difference in the expression levels of zinc transporters ZIPs and ZnTs between the two types of astrocytes, while expression of zinc releasable channel TRPM7 was found on the plasma membrane in ddY-astrocytes, but not in C57BL/6-astrocytes, although the total cellular expression levels were almost the same. In addition, a TRPM7 blocker, 2-aminoethoxydiphenyl borate, increased the intracellular zinc level in H2O2-treated ddY-, but not C57BL/6-astrocytes. Collectively, it is suggested that vulnerability of astrocytes to oxidative stress depends on an increased level of intracellular labile zinc, and TRPM7 localized on the plasma membrane contributes, at least in part, to ameliorate the cell injury by decreasing the zinc level.
Collapse
Affiliation(s)
- Takahiro Furuta
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-84114, Japan
| | - Akihiro Ohishi
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-84114, Japan
| | - Kazuki Nagasawa
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-84114, Japan.
| |
Collapse
|
34
|
Yan Y, Yu J, Gao Y, Kumar G, Guo M, Zhao Y, Fang Q, Zhang H, Yu J, Jiang Y, Zhang HT, Ma CG. Therapeutic potentials of the Rho kinase inhibitor Fasudil in experimental autoimmune encephalomyelitis and the related mechanisms. Metab Brain Dis 2019; 34:377-384. [PMID: 30552558 DOI: 10.1007/s11011-018-0355-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/25/2018] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS), Parkinson's disease (PD), Alzheimer's disease (AD), and other neurodegenerative diseases of central nervous system (CNS) disorders are serious human health problems. Rho-kinase (ROCK) is emerging as a potentially important therapeutic target relevant to inflammatory neurodegeneration diseases. This is supported by studies showing the beneficial effects of fasudil, a ROCK inhibitor, in inflammatory neurodegeneration diseases. MS is an autoimmune disease resulting from inflammation and demyelination in the white matter of the CNS. It has been postulated that activation of Rho/ROCK causes neuropathological changes accompanied with related clinical symptoms, which are improved by treatment with ROCK inhibitors. Therefore, inhibition of abnormal activation of the Rho/ROCK signaling pathway appears to be a new mechanism for treating CNS diseases. In this review, we extensively discussed the role of ROCK inhibitors, summarized the efficacy of fasudil in the MS conventional animal model of experimental autoimmune encephalomyelitis (EAE), both in vivo and in vitro, and highlighted the mechanism involved. Overall, the findings collected in this review support the role of the ROCK signaling pathway in neurodegenerative diseases. Hence, ROCK inhibitors such as fasudil can be novel, and efficacious treatment for inflammatory neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuqing Yan
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Jiezhong Yu
- Institute of Brain Science, Shanxi Datong University, Datong, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Ye Gao
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Gajendra Kumar
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Hong Kong
| | - Minfang Guo
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Yijin Zhao
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Qingli Fang
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Huiyu Zhang
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Jingwen Yu
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Yuqiang Jiang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
| | - Han-Ting Zhang
- Institute of Brain Science, Shanxi Datong University, Datong, China.
- Departments of Behavioral Medicine & Psychiatry, Physiology & Pharmacology, and Neuroscience, the Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA.
| | - Cun-Gen Ma
- Institute of Brain Science, Shanxi Datong University, Datong, China.
- "2011" Collaborative Innovation Center/Research Center of Neurobiology, Taiyuan, China.
| |
Collapse
|
35
|
Putra M, Sharma S, Gage M, Gasser G, Hinojo-Perez A, Olson A, Gregory-Flores A, Puttachary S, Wang C, Anantharam V, Thippeswamy T. Inducible nitric oxide synthase inhibitor, 1400W, mitigates DFP-induced long-term neurotoxicity in the rat model. Neurobiol Dis 2019; 133:104443. [PMID: 30940499 DOI: 10.1016/j.nbd.2019.03.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/22/2019] [Accepted: 03/28/2019] [Indexed: 11/26/2022] Open
Abstract
Chemical nerve agents (CNA) are increasingly becoming a threat to both civilians and military personnel. CNA-induced acute effects on the nervous system have been known for some time and the long-term consequences are beginning to emerge. In this study, we used diisopropylfluorophosphate (DFP), a seizurogenic CNA to investigate the long-term impact of its acute exposure on the brain and its mitigation by an inducible nitric oxide synthase (iNOS) inhibitor, 1400W as a neuroprotectant in the rat model. Several experimental studies have demonstrated that DFP-induced seizures and/or status epilepticus (SE) causes permanent brain injury, even after the countermeasure medication (atropine, oxime, and diazepam). In the present study, DFP-induced SE caused a significant increase in iNOS and 3-nitrotyrosine (3-NT) at 24 h, 48 h, 7d, and persisted for a long-term (12 weeks post-exposure), which led to the hypothesis that iNOS is a potential therapeutic target in DFP-induced brain injury. To test the hypothesis, we administered 1400W (20 mg/kg, i.m.) or the vehicle twice daily for the first three days of post-exposure. 1400W significantly reduced DFP-induced iNOS and 3-NT upregulation in the hippocampus and piriform cortex, and the serum nitrite levels at 24 h post-exposure. 1400W also prevented DFP-induced mortality in <24 h. The brain immunohistochemistry (IHC) at 7d post-exposure revealed a significant reduction in gliosis and neurodegeneration (NeuN+ FJB positive cells) in the 1400W-treated group. 1400W, in contrast to the vehicle, caused a significant reduction in the epileptiform spiking and spontaneous recurrent seizures (SRS) during 12 weeks of continuous video-EEG study. IHC of brain sections from the same animals revealed a significant reduction in reactive gliosis (both microgliosis and astrogliosis) and neurodegeneration across various brain regions in the 1400W-treated group when compared to the vehicle-treated group. A multiplex assay from hippocampal lysates at 6 weeks post-exposure showed a significant increase in several key pro-inflammatory cytokines/chemokines such as IL-1α, TNFα, IL-1β, IL-2, IL-6, IL-12, IL-17a, MCP-1, LIX, and Eotaxin, and a growth factor, VEGF in the vehicle-treated animals. 1400W significantly suppressed IL-1α, TNFα, IL-2, IL-12, and MCP-1 levels. It also suppressed DFP-induced serum nitrite levels at 6 weeks post-exposure. In the Morris water maze, the vehicle-treated animals spent significantly less time in the target quadrant in a probe trial at 9d post-exposure compared to their time spent in the same quadrant 11 days previously (i.e., 2 days prior to DFP exposure). Such a difference was not observed in the 1400W and control groups. However, learning and short-term memory were unaffected when tested at 10-16d and 28-34d post-exposure. Accelerated rotarod, horizontal bar test, and the forced swim test revealed no significant changes between groups. Overall, the findings from this study suggest that 1400W may be considered as a potential therapeutic agent as a follow-on therapy for CNA exposure, after controlling the acute symptoms, to prevent mortality and some of the long-term neurotoxicity parameters such as epileptiform spiking, SRS, neurodegeneration, reactive gliosis in some brain regions, and certain key proinflammatory cytokines and chemokine.
Collapse
Affiliation(s)
- Marson Putra
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, United States
| | - Shaunik Sharma
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, United States
| | - Meghan Gage
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, United States
| | | | - Andy Hinojo-Perez
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, United States
| | - Ashley Olson
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, United States
| | - Adriana Gregory-Flores
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, United States
| | - Sreekanth Puttachary
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, United States
| | - Chong Wang
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, United States
| | | | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
36
|
Cieślak M, Roszek K, Wujak M. Purinergic implication in amyotrophic lateral sclerosis-from pathological mechanisms to therapeutic perspectives. Purinergic Signal 2019; 15:1-15. [PMID: 30430356 PMCID: PMC6439052 DOI: 10.1007/s11302-018-9633-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/01/2018] [Indexed: 12/22/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a clinically heterogeneous disorder characterized by degeneration of upper motor neurons in the brainstem and lower motor neurons in the spinal cord. Multiple mechanisms of motor neuron injury have been implicated, including more than 20 different genetic factors. The pathogenesis of ALS consists of two stages: an early neuroprotective stage and a later neurotoxic. During early phases of disease progression, the immune system through glial and T cell activities provides anti-inflammatory factors that sustain motor neuron viability. As the disease progresses and motor neuron injury accelerates, a rapidly succeeding neurotoxic phase develops. A well-orchestrated purine-mediated dialog among motor neurons, surrounding glia and immune cells control the beneficial and detrimental activities occurring in the nervous system. In general, low adenosine triphosphate (ATP) concentrations protect cells against excitotoxic stimuli through purinergic P2X4 receptor, whereas high concentrations of ATP trigger toxic P2X7 receptor activation. Finally, adenosine is also involved in ALS progression since A2A receptor antagonists prevent motor neuron death. Given the complex cellular cross-talk occurring in ALS and the recognized function of extracellular nucleotides and adenosine in neuroglia communication, the comprehensive understanding of purinome dynamics might provide new research perspectives to decipher ALS and help to design more efficient and targeted drugs. This review will focus on the purinergic players involved in ALS etiology and disease progression and current therapeutic strategies to enhance neuroprotection and suppress neurotoxicity.
Collapse
Affiliation(s)
- M Cieślak
- Neurology Clinic, Marek Cieślak, Toruń, Poland
| | - K Roszek
- Department of Biochemistry, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Toruń, 1 Lwowska St, 87-100, Toruń, Poland
| | - M Wujak
- Department of Biochemistry, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Toruń, 1 Lwowska St, 87-100, Toruń, Poland.
| |
Collapse
|
37
|
Ueba Y, Aratake T, Onodera KI, Higashi Y, Hamada T, Shimizu T, Shimizu S, Yawata T, Nakamura R, Akizawa T, Ueba T, Saito M. Attenuation of zinc-enhanced inflammatory M1 phenotype of microglia by peridinin protects against short-term spatial-memory impairment following cerebral ischemia in mice. Biochem Biophys Res Commun 2018; 507:476-483. [DOI: 10.1016/j.bbrc.2018.11.067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/12/2018] [Indexed: 01/04/2023]
|
38
|
McAllister BB, Wright DK, Wortman RC, Shultz SR, Dyck RH. Elimination of vesicular zinc alters the behavioural and neuroanatomical effects of social defeat stress in mice. Neurobiol Stress 2018; 9:199-213. [PMID: 30450385 PMCID: PMC6234281 DOI: 10.1016/j.ynstr.2018.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/26/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic stress can have deleterious effects on mental health, increasing the risk of developing depression or anxiety. But not all individuals are equally affected by stress; some are susceptible while others are more resilient. Understanding the mechanisms that lead to these differing outcomes has been a focus of considerable research. One unexplored mechanism is vesicular zinc – zinc that is released by neurons as a neuromodulator. We examined how chronic stress, induced by repeated social defeat, affects mice that lack vesicular zinc due to genetic deletion of zinc transporter 3 (ZnT3). These mice, unlike wild type mice, did not become socially avoidant of a novel conspecific, suggesting resilience to stress. However, they showed enhanced sensitivity to the potentiating effect of stress on cued fear memory. Thus, the contribution of vesicular zinc to stress susceptibility is not straightforward. Stress also increased anxiety-like behaviour but produced no deficits in a spatial Y-maze test. We found no evidence that microglial activation or hippocampal neurogenesis accounted for the differences in behavioural outcome. Volumetric analysis revealed that ZnT3 KO mice have larger corpus callosum and parietal cortex volumes, and that corpus callosum volume was decreased by stress in ZnT3 KO, but not wild type, mice.
Collapse
Key Words
- BLA, Basolateral amygdala
- CC, Corpus callosum
- Chronic stress
- Depression
- EPM, Elevated plus-maze
- Fear memory
- LV, Lateral ventricles
- Magnetic resonance imaging (MRI)
- NAc, Nucleus accumbens
- NSF, Novelty-suppressed feeding
- PBS, Phosphate-buffered saline
- PFA, Paraformaldehyde
- PFC, Prefrontal cortex
- RSD, Repeated social defeat
- SLC30A3
- Synaptic zinc
- ZnT3, Zinc transporter 3
- dHPC, Dorsal hippocampus
- vHPC, Ventral hippocampus
Collapse
Affiliation(s)
- Brendan B McAllister
- Department of Psychology & Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - David K Wright
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Ryan C Wortman
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Richard H Dyck
- Department of Psychology & Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
39
|
Aratake T, Higashi Y, Ueba Y, Hamada T, Shimizu T, Shimizu S, Yawata T, Ueba T, Saito M. The inhibitory role of intracellular free zinc in the regulation of Arg-1 expression in interleukin-4-induced activation of M2 microglia. Metallomics 2018; 10:1501-1509. [PMID: 30206632 DOI: 10.1039/c8mt00248g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Microglia, the resident immune cells of the central nervous system, can display a pro-inflammatory M1 phenotype or an anti-inflammatory M2 phenotype. Arginase (Arg)-1 expressed in interleukin-4 (IL-4)-induced M2 microglia reduces nitric oxide (NO) production by competing with inducible NO synthase for l-arginine, which contributes to the attenuation of brain inflammation. Although previous studies have indicated that brain zinc promotes M1 activation, the effect of zinc on M2 microglial activation remains to be determined. In the present study, murine primary microglia treated with 10 ng mL-1 IL-4 exhibited increased Arg-1 mRNA expression and levels of intracellular free zinc. Chelation of this increased intracellular free zinc by the cell permeable zinc chelator N,N,N',N'-tetrakis-(2-pyridylmethyl)ethylenediamine (TPEN) aggravated the IL-4-induced mRNA expression and enzymatic activity of Arg-1. However, the cell impermeable zinc chelator CaEDTA had no effect on Arg-1 expression or cytosolic levels of free zinc in IL-4-induced M2-polarized microglia. Furthermore, treatment with IL-4 resulted in upregulation of phagocytic activity in microglia, while administration of TPEN abolished IL-4-induced phagocytic activity. Moreover, this effect was reversed vial-arginine supplementation. These findings suggest that IL-4 induces an increase in intracellular free zinc in microglia, which may act as a negative regulator of IL-4-induced Arg-1 expression, and that such negative regulation is essential for microglial phagocytic activity.
Collapse
Affiliation(s)
- Takaaki Aratake
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Youichirou Higashi
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Yusuke Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Tomoya Hamada
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Takahiro Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Shogo Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Toshio Yawata
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Tetsuya Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| |
Collapse
|
40
|
Zou S, Shimizu T, Shimizu S, Higashi Y, Nakamura K, Ono H, Aratake T, Saito M. Possible role of hydrogen sulfide as an endogenous relaxation factor in the rat bladder and prostate. Neurourol Urodyn 2018; 37:2519-2526. [DOI: 10.1002/nau.23788] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Suo Zou
- Department of Pharmacology; Kochi Medical School; Kochi University; Nankoku Kochi Japan
| | - Takahiro Shimizu
- Department of Pharmacology; Kochi Medical School; Kochi University; Nankoku Kochi Japan
| | - Shogo Shimizu
- Department of Pharmacology; Kochi Medical School; Kochi University; Nankoku Kochi Japan
| | - Youichirou Higashi
- Department of Pharmacology; Kochi Medical School; Kochi University; Nankoku Kochi Japan
| | - Kumiko Nakamura
- Department of Pharmacology; Kochi Medical School; Kochi University; Nankoku Kochi Japan
| | - Hideaki Ono
- Department of Pharmacology; Kochi Medical School; Kochi University; Nankoku Kochi Japan
| | - Takaaki Aratake
- Department of Pharmacology; Kochi Medical School; Kochi University; Nankoku Kochi Japan
| | - Motoaki Saito
- Department of Pharmacology; Kochi Medical School; Kochi University; Nankoku Kochi Japan
| |
Collapse
|
41
|
Garza-Lombó C, Posadas Y, Quintanar L, Gonsebatt ME, Franco R. Neurotoxicity Linked to Dysfunctional Metal Ion Homeostasis and Xenobiotic Metal Exposure: Redox Signaling and Oxidative Stress. Antioxid Redox Signal 2018; 28:1669-1703. [PMID: 29402131 PMCID: PMC5962337 DOI: 10.1089/ars.2017.7272] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Essential metals such as copper, iron, manganese, and zinc play a role as cofactors in the activity of a wide range of processes involved in cellular homeostasis and survival, as well as during organ and tissue development. Throughout our life span, humans are also exposed to xenobiotic metals from natural and anthropogenic sources, including aluminum, arsenic, cadmium, lead, and mercury. It is well recognized that alterations in the homeostasis of essential metals and an increased environmental/occupational exposure to xenobiotic metals are linked to several neurological disorders, including neurodegeneration and neurodevelopmental alterations. Recent Advances: The redox activity of essential metals is key for neuronal homeostasis and brain function. Alterations in redox homeostasis and signaling are central to the pathological consequences of dysfunctional metal ion homeostasis and increased exposure to xenobiotic metals. Both redox-active and redox-inactive metals trigger oxidative stress and damage in the central nervous system, and the exact mechanisms involved are starting to become delineated. CRITICAL ISSUES In this review, we aim to appraise the role of essential metals in determining the redox balance in the brain and the mechanisms by which alterations in the homeostasis of essential metals and exposure to xenobiotic metals disturb the cellular redox balance and signaling. We focus on recent literature regarding their transport, metabolism, and mechanisms of toxicity in neural systems. FUTURE DIRECTIONS Delineating the specific mechanisms by which metals alter redox homeostasis is key to understand the pathological processes that convey chronic neuronal dysfunction in neurodegenerative and neurodevelopmental disorders. Antioxid. Redox Signal. 28, 1669-1703.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska.,2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Yanahi Posadas
- 3 Departamentos de Farmacología y de, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México .,4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - Liliana Quintanar
- 4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - María E Gonsebatt
- 2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Rodrigo Franco
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska
| |
Collapse
|
42
|
Wang J, Xing H, Wan L, Jiang X, Wang C, Wu Y. Treatment targets for M2 microglia polarization in ischemic stroke. Biomed Pharmacother 2018; 105:518-525. [PMID: 29883947 DOI: 10.1016/j.biopha.2018.05.143] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023] Open
Abstract
As the first line of defense in the nervous system, resident microglia are the predominant immune cells in the brain. In diseases of the central nervous system such as stroke, Alzheimer's disease, and Parkinson's disease, they often cause inflammation or phagocytosis; however, some studies have found that despite the current controversy over M1, M2 polarization could be beneficial. Ischemic stroke is the third most common cause of death in humans. Patients who survive an ischemic stroke might experience a clear decline in their quality of life, owing to conditions such as hemiplegic paralysis and aphasia. After stroke, the activated microglia become a double-edged sword, with distinct phenotypic changes to the deleterious M1 and neuroprotective M2 types. Therefore, methods for promoting the differentiation of microglia into the M2 polarized form to alleviate harmful reactions after stroke have become a topic of interest in recent years. Subsequently, the discovery of new drugs related to M2 polarization has enabled the realization of targeted therapies. In the present review, we discussed the neuroprotective effects of microglia M2 polarization and the potential mechanisms and drugs by which microglia can be transformed into the M2 polarized type after stroke.
Collapse
Affiliation(s)
- Ji Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongyi Xing
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Wan
- The Children's Hospital of Soochow, Jiangsu, Hematology and Oncology, China
| | - Xingjun Jiang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chen Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
43
|
Tronel C, Largeau B, Santiago Ribeiro MJ, Guilloteau D, Dupont AC, Arlicot N. Molecular Targets for PET Imaging of Activated Microglia: The Current Situation and Future Expectations. Int J Mol Sci 2017; 18:ijms18040802. [PMID: 28398245 PMCID: PMC5412386 DOI: 10.3390/ijms18040802] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/15/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022] Open
Abstract
Microglia, as cellular mediators of neuroinflammation, are implicated in the pathogenesis of a wide range of neurodegenerative diseases. Positron emission tomography (PET) imaging of microglia has matured over the last 20 years, through the development of radiopharmaceuticals targeting several molecular biomarkers of microglial activation and, among these, mainly the translocator protein-18 kDa (TSPO). Nevertheless, current limitations of TSPO as a PET microglial biomarker exist, such as low brain density, even in a neurodegenerative setting, expression by other cells than the microglia (astrocytes, peripheral macrophages in the case of blood brain barrier breakdown), genetic polymorphism, inducing a variation for most of TSPO PET radiopharmaceuticals’ binding affinity, or similar expression in activated microglia regardless of its polarization (pro- or anti-inflammatory state), and these limitations narrow its potential interest. We overview alternative molecular targets, for which dedicated radiopharmaceuticals have been proposed, including receptors (purinergic receptors P2X7, cannabinoid receptors, α7 and α4β2 nicotinic acetylcholine receptors, adenosine 2A receptor, folate receptor β) and enzymes (cyclooxygenase, nitric oxide synthase, matrix metalloproteinase, β-glucuronidase, and enzymes of the kynurenine pathway), with a particular focus on their respective contribution for the understanding of microglial involvement in neurodegenerative diseases. We discuss opportunities for these potential molecular targets for PET imaging regarding their selectivity for microglia expression and polarization, in relation to the mechanisms by which microglia actively participate in both toxic and neuroprotective actions in brain diseases, and then take into account current clinicians’ expectations.
Collapse
Affiliation(s)
- Claire Tronel
- INSERM U930, Université François Rabelais de Tours, 10 boulevard Tonnelé, 37032 Tours, France.
| | | | - Maria Joao Santiago Ribeiro
- INSERM U930, Université François Rabelais de Tours, 10 boulevard Tonnelé, 37032 Tours, France.
- CHRU de Tours, 37044 Tours, France.
| | - Denis Guilloteau
- INSERM U930, Université François Rabelais de Tours, 10 boulevard Tonnelé, 37032 Tours, France.
- CHRU de Tours, 37044 Tours, France.
| | - Anne-Claire Dupont
- INSERM U930, Université François Rabelais de Tours, 10 boulevard Tonnelé, 37032 Tours, France.
- CHRU de Tours, 37044 Tours, France.
| | - Nicolas Arlicot
- INSERM U930, Université François Rabelais de Tours, 10 boulevard Tonnelé, 37032 Tours, France.
- CHRU de Tours, 37044 Tours, France.
| |
Collapse
|
44
|
Furuta T, Mukai A, Ohishi A, Nishida K, Nagasawa K. Oxidative stress-induced increase of intracellular zinc in astrocytes decreases their functional expression of P2X7 receptors and engulfing activity. Metallomics 2017; 9:1839-1851. [DOI: 10.1039/c7mt00257b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Exposure of astrocytes to oxidative stress induces an increase of intracellular labile zinc and a decrease of functional expression of P2X7 receptorviaits translocation from the plasma membrane to the cytosol by altering the expression profile of P2X7 receptor and its splice variants, leading to a decrease of their engulfing activity.
Collapse
Affiliation(s)
- Takahiro Furuta
- Department of Environmental Biochemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| | - Ayumi Mukai
- Department of Environmental Biochemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| | - Akihiro Ohishi
- Department of Environmental Biochemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| | - Kentaro Nishida
- Department of Environmental Biochemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| | - Kazuki Nagasawa
- Department of Environmental Biochemistry
- Kyoto Pharmaceutical University
- Yamashina-ku
- Japan
| |
Collapse
|