1
|
Ren ZL, Zhang CX, Zheng YX, Chen CA, Dan-Chen, Lan X, Yan X, Liu Y, He YH, Cheng JL, Han JH, Wang QG, Wang XQ, Cheng FF, Li CX. Refined qingkailing attenuates reactive astrocytes and glial scar formation after ischemia stroke via the EGFR/PLCγ pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156696. [PMID: 40393215 DOI: 10.1016/j.phymed.2025.156696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Ischemic stroke (IS) is the main cause of disability worldwide, and glial scar can impair neurological recovery during the post-stroke period. Refined qingkailing (RQKL) has been demonstrated to be neuroprotective after IS. PURPOSE The purpose of our study was to investigate the effect of RQKL on glial scar after IS. METHODS In this work, rats were used as the model subjects for middle cerebral artery occlusion (MCAO), with 7 and 14 days serving as the critical observational intervals. The treatments of oxygen and glucose deprivation/reoxygenation (OGD/R) were applied to primary astrocytes and an astrocyte-neuron co-culture model. RESULTS RQKL was effective in improving neurological dysfunction, brain histopathologic manifestations, and reducing the degree of brain atrophy at different stages of glial scar. It also decreased the expression of glial fibrillary acidic protein (GFAP), neurocan, and brevican, and increased the expression of microtubule associated protein 2 (MAP2). In primary astrocyte culture, RQKL reduced the activation and proliferation of astrocytes. In an astrocyte and neuron co-culture model, RQKL decreased the expression of GFAP and brevican in astrocytes, and increased the expression of MAP2 and NF200 in neurons. Epidermal growth factor receptor (EGFR) and p-PLCγ expression was strongly increased following IS, according to both in vivo and in vitro tests, while RQKL decreased EGFR and p-PLCγ expression. CONCLUSION When considered collectively, these findings imply that the EGFR/PLCγ signaling pathway is crucial for the activation of astrocytes and the formation of glial scars following IS. Also, RQKL affects neurons by blocking the EGFR/PLCγ signaling pathway on astrocytes, which diminishes the activation of astrocytes and the development of glial scars.
Collapse
Affiliation(s)
- Zi-Lin Ren
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China
| | - Chu-Xin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China
| | - Yu-Xiao Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China
| | - Cong-Ai Chen
- Beijing Chinese Medicine Hospital, Capital Medical University, Beijing 100010, China
| | - Dan-Chen
- Chongqing College of Traditional Chinese Medicine, Chongqing 402760, China
| | - Xin Lan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China
| | - Xin Yan
- National Narcotics Laboratory Beijing Regional Center, Beijing 100164, China
| | - Ying Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China
| | - Yan-Hui He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China
| | - Jia-Lin Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China
| | - Jin-Hua Han
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China
| | - Qing-Guo Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China
| | - Xue-Qian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China.
| | - Fa-Feng Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China.
| | - Chang-Xiang Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11 North Third Ring Road East, Beijing 100029, China.
| |
Collapse
|
2
|
Min E, Ko MY, Kim M, Park H, Kim Y, Kim KK, Lee BS, Hyun SA, Ka M. Perfluorooctanoic acid (PFOA) activates astrogliosis-associated neuroinflammation through ER stress-autophagy axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 979:179546. [PMID: 40288163 DOI: 10.1016/j.scitotenv.2025.179546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Perfluorooctanoic acid (PFOA), a perfluoroalkyl acid, induces neuroinflammation. However, present understanding regarding its fundamental role in neuroinflammation remains limited. Therefore, in this study, we aimed to clarify the potential association between PFOA and astrocyte activation via the modulation of the endoplasmic reticulum (ER) stress-autophagy axis. The results obtained revealed that PFOA activated astroglia in A-172 astrocytoma cells and primary astrocytes by upregulating the expression levels of autophagy-related proteins (ATG5, BECN1, SQSTM1, and MAP1LC3B-II). It also activated autophagy in A-172 astrocytoma cells and primary astrocytes via the upstream activation of ER stress-related proteins, such as ATF4, GRP78, and CHOP. Further, the pharmacological inhibition of ER stress as well as autophagy prevented PFOA-induced activation of astrogliosis in PFOA-treated A-172 cells and primary astrocytes. We also observed that PFOA-mediated activation of GFAP upregulated the transcription of pro-inflammatory cytokines, such as IL-1β, TNF-α, and IL-6. These findings confirmed the existence of a relationship between ER stress-induced autophagy and astrogliosis in PFOA-treated astrocytes, suggesting that targeting the ER stress-autophagy axis may be a potential therapeutic strategy for reducing PFOA-induced neuroinflammation.
Collapse
Affiliation(s)
- Euijun Min
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Moon Yi Ko
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Minjeong Kim
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Heejin Park
- Center for Toxicologic Pathology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Younhee Kim
- Center for Toxicologic Pathology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Kee K Kim
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Byoung-Seok Lee
- Center for Toxicologic Pathology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| | - Sung-Ae Hyun
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| | - Minhan Ka
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34114, Republic of Korea.
| |
Collapse
|
3
|
Salem MA, Khalil HMA, Manaa EG, Bass AKA, Osama N, Samaka RM, Ibrahim MT, Hamdan DI. Antioxidant Potential of Selected Apiaceae Plant Extracts: A Study Focused on the Chemical Composition and Neuroprotective Effect of Coriandrum sativum L. Extract Against Lead (Pb)-Induced Neurotoxicity in Rats. Biol Trace Elem Res 2025:10.1007/s12011-025-04627-9. [PMID: 40261543 DOI: 10.1007/s12011-025-04627-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
Lead is a common environmental pollutant. It can affect several body systems including the central nervous system (CNS). Lead can disrupt the nervous system by different mechanisms including oxidative stress, inflammation, disruption of neurotransmission, and aberrant autophagy. Apiaceous species have been used traditionally as food flavoring and medicine, representing a rich source of bioactive compounds. In the current study, the antioxidant power of four Apiaceous extracts (Foeniculum vulgare L., Pimpinella anisum L., Coriandrum sativum L., and Cuminum cyminum L.) was evaluated. Additionally, the metabolite profiles of the selected species were comprehensively investigated by untargeted liquid chromatography electrospray ionization tandem mass spectrometry (LC/ESI-MS/MS) coupled to chemometry. Coriander (Coriandrum sativum L.) extract showed the highest radical scavenging activity and reducing power. Coriander was further subjected to in vivo evaluation of its protective effect against Lead (Pb)-induced neurotoxicity. Administration of coriander extracts improved the short- and long-term memory performance and decreased hippocampal Pb content in Pb-intoxicated rats. Moreover, it attenuated hippocampal oxidative stress, neurochemical changes, and exhibited anti-inflammatory effect in the hippocampal tissue. Further, coriander extracts attenuated Pb inhibitory effect on the mammalian target of Rapamycin (mTORC1) pathway resulting in upregulation of Phospho-p70 S6 Kinase (P-P70S6K) and Phospho-S6 Ribosomal Protein (PS6) and downregulation of Beclin-1. Additionally, some selected coriander ingredients were subjected to molecular docking to examine their regulatory effect on mTORC-1 and IκB kinase complex (Ikk-β). The present findings highlight the future pharmaceutical utilization of coriander extract as valuable source of phenolic compounds that can be used as antioxidant, anti-inflammatory, and neuroprotective agents against Pb-induced neurotoxicity.
Collapse
Affiliation(s)
- Mohamed A Salem
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia University, Gamal Abd El Nasr St., Shibin Elkom, Menoufia, 32511, Egypt
- The BioActives Lab, Biological and Environment Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Heba M A Khalil
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
- Faculty of Veterinary medicine, King Salman International University, South Sinai, Ras Sudr, Egypt
| | - Eman G Manaa
- Department of Pharmacognosy, Faculty of Pharmacy, Alsalam University, Al Gharbiyah, Egypt
| | - Amr K A Bass
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Menoufia University, Menoufia, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Menoufia National University, Km Cairo-Alexandria Agricultural Road, Menofia, Egypt
| | - Nada Osama
- Biochemistry Department, Faculty of Pharmacy, Menoufia University, Gamal Abd El Nasr St., Shibin Elkom, Menoufia, 32511, Egypt.
| | - Rehab M Samaka
- Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Magda T Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, 11785, Egypt
| | - Dalia I Hamdan
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia University, Gamal Abd El Nasr St., Shibin Elkom, Menoufia, 32511, Egypt
- Department of Pharmacognosy and Natural Products, Faculty of Pharmacy, Menoufia National University, Km Cairo-Alexandria Agricultural Road, Menofia, Egypt
| |
Collapse
|
4
|
Kwon D, Kim Y, Cho SH. Antidepressant Effects of Ginsenoside Rc on L-Alpha-Aminoadipic Acid-Induced Astrocytic Ablation and Neuroinflammation in Mice. Int J Mol Sci 2024; 25:9673. [PMID: 39273621 PMCID: PMC11396248 DOI: 10.3390/ijms25179673] [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: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Depression is a prevalent and debilitating mental disorder that affects millions worldwide. Current treatments, such as antidepressants targeting the serotonergic system, have limitations, including delayed onset of action and high rates of treatment resistance, necessitating novel therapeutic strategies. Ginsenoside Rc (G-Rc) has shown potential anti-inflammatory and neuroprotective effects, but its antidepressant properties remain unexplored. This study investigated the antidepressant effects of G-Rc in an L-alpha-aminoadipic acid (L-AAA)-induced mouse model of depression, which mimics the astrocytic pathology and neuroinflammation observed in major depressive disorder. Mice were administered G-Rc, vehicle, or imipramine orally after L-AAA injection into the prefrontal cortex. G-Rc significantly reduced the immobility time in forced swimming and tail suspension tests compared to vehicle treatment, with more pronounced effects than imipramine. It also attenuated the expression of pro-inflammatory cytokines (TNF-α, IL-6, TGF-β, lipocalin-2) and alleviated astrocytic degeneration, as indicated by increased GFAP and decreased IBA-1 levels. Additionally, G-Rc modulated apoptosis-related proteins, decreasing caspase-3 and increasing Bcl-2 levels compared to the L-AAA-treated group. These findings suggest that G-Rc exerts antidepressant effects by regulating neuroinflammation, astrocyte-microglia crosstalk, and apoptotic pathways in the prefrontal cortex, highlighting its potential as a novel therapeutic agent for depression.
Collapse
Affiliation(s)
- Dohyung Kwon
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yunna Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung-Hun Cho
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Neuropsychiatry of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
- Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
5
|
Pereira LTG, Vilela WR, Bellozi PMQ, Engel DF, de Paula GC, de Andrade RR, Mortari MR, de Melo Teixeira M, Coleine C, Figueiredo CP, de Bem AF, Amato AA. Fecal microbiota transplantation ameliorates high-fat diet-induced memory impairment in mice. J Neurochem 2024; 168:2893-2907. [PMID: 38934224 DOI: 10.1111/jnc.16156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
Gut dysbiosis is linked to metabolic and neurodegenerative diseases and comprises a plausible link between high-fat diet (HFD) and brain dysfunction. Here we show that gut microbiota modulation by either antibiotic treatment for 5 weeks or a brief 3-day fecal microbiota transplantation (FMT) regimen from low-fat (control) diet-fed mice decreased weight gain, adipose tissue hypertrophy, and glucose intolerance induced by HFD in C57BL/6 male mice. Notably, gut microbiota modulation by FMT completely reversed impaired recognition memory induced by HFD, whereas modulation by antibiotics had less pronounced effect. Improvement in recognition memory by FMT was accompanied by decreased HFD-induced astrogliosis in the hippocampal cornu ammonis region. Gut microbiome composition analysis indicated that HFD diminished microbiota diversity compared to control diet, whereas FMT partially restored the phyla diversity. Our findings reinforce the role of the gut microbiota on HFD-induced cognitive impairment and suggest that modulating the gut microbiota may be an effective strategy to prevent metabolic and cognitive dysfunction associated with unfavorable dietary patterns.
Collapse
Affiliation(s)
| | - Wembley Rodrigues Vilela
- Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Paula Maria Quaglio Bellozi
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
- Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Daiane Fátima Engel
- School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | | | | | - Márcia Renata Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Biology Institute, University of Brasilia, Federal District, Brazil
| | | | - Claudia Coleine
- Department of Ecological and Biological Sciences, University of Tuscia, Viterbo, Italy
| | - Cláudia Pinto Figueiredo
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andreza Fabro de Bem
- Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brasilia, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Angélica Amorim Amato
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
6
|
Sun Y, Cai H, Yang D, Yu N, Sun L, Xu J, Yuan H, Yang R, Song L, Liu H, Ma C, Liu Z. β-arrestin2 is indispensable for the antidepressant effects of fluoxetine via inhibiting astrocytic pyroptosis in chronic mild stress mouse model for depression. Eur J Pharmacol 2024; 976:176693. [PMID: 38834095 DOI: 10.1016/j.ejphar.2024.176693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
β-arrestin2 is a versatile protein for signaling transduction in brain physiology and pathology. Herein, we investigated the involvement of β-arrestin2 in pharmacological effects of fluoxetine for depression. A chronic mild stress (CMS) model was established using wild-type (WT) and β-arrestin2-/- mice. Behavioral results demonstrated that CMS mice showed increased immobility time in the tail suspension test and forced swimming test, elevated concentrations of pro-inflammatory factors in peripheral blood, increased expression of pyroptosis-related proteins, and increased co-labeling of glial fibrillary acidic protein and Caspase1 p10 in the hippocampus compared to the CON group. Treatment with fluoxetine (FLX) ameliorated these conditions. However, compared with the β-arrestin2-/- CMS group, these results of the β-arrestin2-/- CMS + FLX group showed no significant changes. These results suggested that the above effects of FLX could be eliminated by knocking out β-arrestin2. Mass spectrometry implying that FLX promoted the binding of β-arrestin2 to the NLRP2 inflammasome of depressed mice. Subsequently, the results of the cellular experiments suggested that the 5HT2B receptor antagonist may attenuate L-kynurenine + ATP-induced cell pyroptosis by attenuating NLRP2 binding to β-arrestin2. We further found that the lack of β-arrestin2 eliminated the anti-pyroptosis effect of fluoxetine. In conclusion, β-arrestin2 is an essential protein for fluoxetine to alleviate pyroptosis in the hippocampal astrocytes of CMS mice. Mechanistically, we found that the 5-HT2BR-β-arrestin2-NLRP2 axis is vital for maintaining the antidepressant effects of fluoxetine.
Collapse
Affiliation(s)
- Yiming Sun
- The First Affiliated Hospital of Bengbu Medical University, Changhuai Road, Bengbu, 233000, Anhui, China; School of Pharmacy, Bengbu Medical University, Donghhai Avenue, Bengbu, 233030, Anhui, China
| | - Hui Cai
- The First Affiliated Hospital of Bengbu Medical University, Changhuai Road, Bengbu, 233000, Anhui, China
| | - Daofeng Yang
- The First Affiliated Hospital of Bengbu Medical University, Changhuai Road, Bengbu, 233000, Anhui, China
| | - Nengyi Yu
- School of Pharmacy, Bengbu Medical University, Donghhai Avenue, Bengbu, 233030, Anhui, China
| | - Lejie Sun
- School of Pharmacy, Bengbu Medical University, Donghhai Avenue, Bengbu, 233030, Anhui, China
| | - Jingxuan Xu
- School of Pharmacy, Bengbu Medical University, Donghhai Avenue, Bengbu, 233030, Anhui, China
| | - Hongwei Yuan
- School of Pharmacy, Bengbu Medical University, Donghhai Avenue, Bengbu, 233030, Anhui, China
| | - Rong Yang
- School of Pharmacy, Bengbu Medical University, Donghhai Avenue, Bengbu, 233030, Anhui, China
| | - Lele Song
- The First Affiliated Hospital of Bengbu Medical University, Changhuai Road, Bengbu, 233000, Anhui, China
| | - Hao Liu
- School of Pharmacy, Bengbu Medical University, Donghhai Avenue, Bengbu, 233030, Anhui, China
| | - Chengyao Ma
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| | - Zhe Liu
- The First Affiliated Hospital of Bengbu Medical University, Changhuai Road, Bengbu, 233000, Anhui, China; School of Pharmacy, Bengbu Medical University, Donghhai Avenue, Bengbu, 233030, Anhui, China.
| |
Collapse
|
7
|
League AF, Yadav-Samudrala BJ, Kolagani R, Cline CA, Jacobs IR, Manke J, Niphakis MJ, Cravatt BF, Lichtman AH, Ignatowska-Jankowska BM, Fitting S. A helping HAND: therapeutic potential of MAGL inhibition against HIV-1-associated neuroinflammation. Front Immunol 2024; 15:1374301. [PMID: 38835765 PMCID: PMC11148243 DOI: 10.3389/fimmu.2024.1374301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/25/2024] [Indexed: 06/06/2024] Open
Abstract
Background Human immunodeficiency virus (HIV) affects nearly 40 million people globally, with roughly 80% of all people living with HIV receiving antiretroviral therapy. Antiretroviral treatment suppresses viral load in peripheral tissues but does not effectively penetrate the blood-brain barrier. Thus, viral reservoirs persist in the central nervous system and continue to produce low levels of inflammatory factors and early viral proteins, including the transactivator of transcription (Tat). HIV Tat is known to contribute to chronic neuroinflammation and synaptodendritic damage, which is associated with the development of cognitive, motor, and/or mood problems, collectively known as HIV-associated neurocognitive disorders (HAND). Cannabinoid anti-inflammatory effects are well documented, but therapeutic utility of cannabis remains limited due to its psychotropic effects, including alterations within brain regions encoding reward processing and motivation, such as the nucleus accumbens. Alternatively, inhibiting monoacylglycerol lipase (MAGL) has demonstrated therapeutic potential through interactions with the endocannabinoid system. Methods The present study utilized a reward-related operant behavioral task to quantify motivated behavior in female Tat transgenic mice treated with vehicle or MAGL inhibitor MJN110 (1 mg/kg). Brain tissue was collected to assess dendritic injury and neuroinflammatory profiles, including dendritic microtubule-associated protein (MAP2ab) intensity, microglia density, microglia morphology, astrocyte density, astrocytic interleukin-1ß (IL-1ß) colocalization, and various lipid mediators. Results No significant behavioral differences were observed; however, MJN110 protected against Tat-induced dendritic injury by significantly upregulating MAP2ab intensity in the nucleus accumbens and in the infralimbic cortex of Tat(+) mice. No or only minor effects were noted for Iba-1+ microglia density and/or microglia morphology. Further, Tat increased GFAP+ astrocyte density in the infralimbic cortex and GFAP+ astrocytic IL-1ß colocalization in the nucleus accumbens, with MJN110 significantly reducing these measures in Tat(+) subjects. Lastly, selected HETE-related inflammatory lipid mediators in the striatum were downregulated by chronic MJN110 treatment. Conclusions These findings demonstrate anti-inflammatory and neuroprotective properties of MJN110 without cannabimimetic behavioral effects and suggest a promising alternative to cannabis for managing neuroinflammation.
Collapse
Affiliation(s)
- Alexis F. League
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Barkha J. Yadav-Samudrala
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ramya Kolagani
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Calista A. Cline
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ian R. Jacobs
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jonathan Manke
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Micah J. Niphakis
- Department of Chemistry, Scripps Research, La Jolla, CA, United States
| | | | - Aron H. Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Sylvia Fitting
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
8
|
Cogut V, Goris M, Jansma A, van der Staaij M, Henning RH. Hippocampal neuroimmune response in mice undergoing serial daily torpor induced by calorie restriction. Front Neuroanat 2024; 18:1334206. [PMID: 38686173 PMCID: PMC11056553 DOI: 10.3389/fnana.2024.1334206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
Hibernating animals demonstrate a remarkable ability to withstand extreme physiological brain changes without triggering adverse neuroinflammatory responses. While hibernators may offer valuable insights into the neuroprotective mechanisms inherent to hibernation, studies using such species are constrained by the limited availability of molecular tools. Laboratory mice may serve as an alternative, entering states of hypometabolism and hypothermia similar to the torpor observed in hibernation when faced with energy shortage. Notably, prolonged calorie restriction (CR) induces serial daily torpor patterns in mice, comparable to species that utilize daily hibernation. Here, we examined the neuroinflammatory response in the hippocampus of male C57BL/6 mice undergoing serial daily torpor induced by a 30% CR for 4 weeks. During daily torpor episodes, CR mice exhibited transient increases in TNF-α mRNA expression, which normalized upon arousal. Concurrently, the CA1 region of the hippocampus showed persistent morphological changes in microglia, characterized by reduced cell branching, decreased cell complexity and altered shape. Importantly, these morphological changes were not accompanied by evident signs of astrogliosis or oxidative stress, typically associated with detrimental neuroinflammation. Collectively, the adaptive nature of the brain's inflammatory response to CR-induced torpor in mice parallels observations in hibernators, highlighting its value for studying the mechanisms of brain resilience during torpor. Such insights could pave the way for novel therapeutic interventions in stroke and neurodegenerative disorders in humans.
Collapse
Affiliation(s)
- Valeria Cogut
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
| | | | | | | | | |
Collapse
|
9
|
Sarkar SK, Willson AML, Jordan MA. The Plasticity of Immune Cell Response Complicates Dissecting the Underlying Pathology of Multiple Sclerosis. J Immunol Res 2024; 2024:5383099. [PMID: 38213874 PMCID: PMC10783990 DOI: 10.1155/2024/5383099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative autoimmune disease characterized by the destruction of the myelin sheath of the neuronal axon in the central nervous system. Many risk factors, including environmental, epigenetic, genetic, and lifestyle factors, are responsible for the development of MS. It has long been thought that only adaptive immune cells, especially autoreactive T cells, are responsible for the pathophysiology; however, recent evidence has indicated that innate immune cells are also highly involved in disease initiation and progression. Here, we compile the available data regarding the role immune cells play in MS, drawn from both human and animal research. While T and B lymphocytes, chiefly enhance MS pathology, regulatory T cells (Tregs) may serve a more protective role, as can B cells, depending on context and location. Cells chiefly involved in innate immunity, including macrophages, microglia, astrocytes, dendritic cells, natural killer (NK) cells, eosinophils, and mast cells, play varied roles. In addition, there is evidence regarding the involvement of innate-like immune cells, such as γδ T cells, NKT cells, MAIT cells, and innate-like B cells as crucial contributors to MS pathophysiology. It is unclear which of these cell subsets are involved in the onset or progression of disease or in protective mechanisms due to their plastic nature, which can change their properties and functions depending on microenvironmental exposure and the response of neural networks in damage control. This highlights the need for a multipronged approach, combining stringently designed clinical data with carefully controlled in vitro and in vivo research findings, to identify the underlying mechanisms so that more effective therapeutics can be developed.
Collapse
Affiliation(s)
- Sujan Kumar Sarkar
- Department of Anatomy, Histology and Physiology, Faculty of Animal Science and Veterinary Medicine, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh
| | - Annie M. L. Willson
- Biomedical Sciences and Molecular Biology, CPHMVS, James Cook University, Townsville, Queensland 4811, Australia
| | - Margaret A. Jordan
- Biomedical Sciences and Molecular Biology, CPHMVS, James Cook University, Townsville, Queensland 4811, Australia
| |
Collapse
|
10
|
Shin HJ, Choi SG, Qu F, Yi MH, Lee CH, Kim SR, Kim HG, Beom J, Yi Y, Kim DK, Joe EH, Song HJ, Kim Y, Kim DW. Peptide-mediated targeted delivery of SOX9 nanoparticles into astrocytes ameliorates ischemic brain injury. NANOSCALE 2024; 16:833-847. [PMID: 38093712 DOI: 10.1039/d3nr01318a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Astrocytes are highly activated following brain injuries, and their activation influences neuronal survival. Additionally, SOX9 expression is known to increase in reactive astrocytes. However, the role of SOX9 in activated astrocytes following ischemic brain damage has not been clearly elucidated yet. Therefore, in the present study, we investigated the role of SOX9 in reactive astrocytes using a poly-lactic-co-glycolic acid (PLGA) nanoparticle plasmid delivery system in a photothrombotic stroke animal model. We designed PLGA nanoparticles to exclusively enhance SOX9 gene expression in glial fibrillary acidic protein (GFAP)-immunoreactive astrocytes. Our observations indicate that PLGA nanoparticles encapsulated with GFAP:SOX9:tdTOM reduce ischemia-induced neurological deficits and infarct volume through the prostaglandin D2 pathway. Thus, the astrocyte-targeting PLGA nanoparticle plasmid delivery system provides a potential opportunity for stroke treatment. Since the only effective treatment currently available is reinstating the blood supply, cell-specific gene therapy using PLGA nanoparticles will open a new therapeutic paradigm for brain injury patients in the future.
Collapse
Affiliation(s)
- Hyo Jung Shin
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon 35015, Korea
- Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| | - Seung Gyu Choi
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon 35015, Korea
| | - Fengrui Qu
- Department of Chemistry and Biochemistry, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea
| | - Hyeong-Geug Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jaewon Beom
- Department of Rehabilitation Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Yoonyoung Yi
- Department of Pediatrics, College of Medicine, Hallym University and Gangdong Sacred Heart Hospital, Seoul 05355, Korea
| | - Do Kyung Kim
- Department of Anatomy, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Eun-Hye Joe
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Hee-Jung Song
- Department of Neurology, Chungnam National University and Sejong Hospital, Sejong 30099, Korea
| | - Yonghyun Kim
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA.
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon 35015, Korea
- Brain Research Institute, Chungnam National University, Daejeon 35015, Korea
| |
Collapse
|
11
|
Andersen AM, Kaalund SS, Marner L, Salvesen L, Pakkenberg B, Olesen MV. Quantitative cellular changes in multiple system atrophy brains. Neuropathol Appl Neurobiol 2023; 49:e12941. [PMID: 37812040 DOI: 10.1111/nan.12941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/21/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
Multiple system atrophy (MSA) is a neurodegenerative disorder characterised by a combined symptomatology of parkinsonism, cerebellar ataxia, autonomic failure and corticospinal dysfunction. In brains of MSA patients, the hallmark lesion is the aggregation of misfolded alpha-synuclein in oligodendrocytes. Even though the underlying pathological mechanisms remain poorly understood, the evidence suggests that alpha-synuclein aggregation in oligodendrocytes may contribute to the neurodegeneration seen in MSA. The primary aim of this review is to summarise the published stereological data on the total number of neurons and glial cell subtypes (oligodendrocytes, astrocytes and microglia) and volumes in brains from MSA patients. Thus, we include in this review exclusively the reports of unbiased quantitative data from brain regions including the neocortex, nuclei of the cerebrum, the brainstem and the cerebellum. Furthermore, we compare and discuss the stereological results in the context of imaging findings and MSA symptomatology. In general, the stereological results agree with the common neuropathological findings of neurodegeneration and gliosis in brains from MSA patients and support a major loss of nigrostriatal neurons in MSA patients with predominant parkinsonism (MSA-P), as well as olivopontocerebellar atrophy in MSA patients with predominant cerebellar ataxia (MSA-C). Surprisingly, the reports indicate only a minor loss of oligodendrocytes in sub-cortical regions of the cerebrum (glial cells not studied in the cerebellum) and negligible changes in brain volumes. In the past decades, the use of stereological methods has provided a vast amount of accurate information on cell numbers and volumes in the brains of MSA patients. Combining different techniques such as stereology and diagnostic imaging (e.g. MRI, PET and SPECT) with clinical data allows for a more detailed interdisciplinary understanding of the disease and illuminates the relationship between neuropathological changes and MSA symptomatology.
Collapse
Affiliation(s)
- Alberte M Andersen
- Centre for Neuroscience and Stereology, Department of Neurology, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
| | - Sanne S Kaalund
- Centre for Neuroscience and Stereology, Department of Neurology, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
| | - Lisbeth Marner
- Department of Clinical Physiology and Nuclear Medicine, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisette Salvesen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
| | - Bente Pakkenberg
- Centre for Neuroscience and Stereology, Department of Neurology, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel V Olesen
- Centre for Neuroscience and Stereology, Department of Neurology, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
| |
Collapse
|
12
|
Denaroso GE, Smith Z, Angeliu CG, Cheli VT, Wang C, Paez PM. Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis. J Neuroinflammation 2023; 20:263. [PMID: 37964385 PMCID: PMC10644533 DOI: 10.1186/s12974-023-02948-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/05/2023] [Indexed: 11/16/2023] Open
Abstract
The experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis was used in combination with a Cav1.2 conditional knock-out mouse (Cav1.2KO) to study the role of astrocytic voltage-gated Ca++ channels in autoimmune CNS inflammation and demyelination. Cav1.2 channels were specifically ablated in Glast-1-positive astrocytes by means of the Cre-lox system before EAE induction. After immunization, motor activity was assessed daily, and a clinical score was given based on the severity of EAE symptoms. Cav1.2 deletion in astrocytes significantly reduced the severity of the disease. While no changes were found in the day of onset and peak disease severity, EAE mean clinical score was lower in Cav1.2KO animals during the chronic phase of the disease. This corresponded to better performance on the rotarod and increased motor activity in Cav1.2KO mice. Furthermore, decreased numbers of reactive astrocytes, activated microglia, and infiltrating lymphocytes were found in the lumbar section of the spinal cord of Cav1.2KO mice 40 days after immunization. The degree of myelin protein loss and size of demyelinated lesions were also attenuated in Cav1.2KO spinal cords. Similar results were found in EAE animals treated with nimodipine, a Cav1.2 Ca++ channel inhibitor with high affinity to the CNS. Mice injected with nimodipine during the acute and chronic phases of the disease exhibited lower numbers of reactive astrocytes, activated microglial, and infiltrating immune cells, as well as fewer demyelinated lesions in the spinal cord. These changes were correlated with improved clinical scores and motor performance. In summary, these data suggest that antagonizing Cav1.2 channels in astrocytes during EAE alleviates neuroinflammation and protects the spinal cord from autoimmune demyelination.
Collapse
Affiliation(s)
- G E Denaroso
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - Z Smith
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - C G Angeliu
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - V T Cheli
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - C Wang
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - P M Paez
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA.
| |
Collapse
|
13
|
Zamani N, Osgoei LT, Aliaghaei A, Zamani N, Hassanian-Moghaddam H. Chronic exposure to methadone induces activated microglia and astrocyte and cell death in the cerebellum of adult male rats. Metab Brain Dis 2023; 38:323-338. [PMID: 36287354 DOI: 10.1007/s11011-022-01108-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 02/03/2023]
Abstract
Methadone is a centrally-acting synthetic opioid analgesic widely used in the methadone maintenance therapy (MMT) programs throughout the world. Considering its neurotoxic effects particularly on the cerebellum, this study aims to address the behavioral and histological alterations in the cerebellar cortex associated with methadone administration. Twenty-four adult male albino rats were randomized into two groups of control and methadone treatment. Methadone was subcutaneously administered (2.5-10 mg/kg) once a day for two consecutive weeks. The functional and structural changes in the cerebellum were compared to the control group. Our data revealed that treating rats with methadone not only induced cerebellar atrophy, but also prompted the actuation of microgliosis, astrogliosis, and apoptotic biomarkers. We further demonstrated that treating rats with methadone increased complexity of astrocyte processes and decreased complexity of microglia processes. Our result showed that methadone impaired motor coordination and locomotor performance and neuromuscular activity. Additionally, relative gene expression of TNF-α, caspase-3 and RIPK3 increased significantly due to methadone. Our findings suggest that methadone administration has a neurodegenerative effect on the cerebellar cortex via dysregulation of microgliosis, astrogliosis, apoptosis, and neuro-inflammation.
Collapse
Affiliation(s)
- Naghmeh Zamani
- Department of Biology, Faculty of Biological Science, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Laya Takbiri Osgoei
- Department of Microbiology, Faculty of Biological Science, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Nasim Zamani
- Department of Clinical Toxicology, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Hassanian-Moghaddam
- Department of Clinical Toxicology, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Haramati A, Rechtman A, Zveik O, Haham N, Brill L, Vaknin-Dembinsky A. IL-6 as a marker for NMOSD disease activity. J Neuroimmunol 2022; 370:577925. [DOI: 10.1016/j.jneuroim.2022.577925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/17/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022]
|
15
|
Tonoli E, Verduci I, Gabrielli M, Prada I, Forcaia G, Coveney C, Savoca MP, Boocock DJ, Sancini G, Mazzanti M, Verderio C, Verderio EAM. Extracellular transglutaminase-2, nude or associated with astrocytic extracellular vesicles, modulates neuronal calcium homeostasis. Prog Neurobiol 2022; 216:102313. [PMID: 35760142 DOI: 10.1016/j.pneurobio.2022.102313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 01/12/2023]
Abstract
We have uncovered a novel role for astrocytes-derived extracellular vesicles (EVs) in controlling intraneuronal Ca2+ concentration ([Ca2+]i) and identified transglutaminase-2 (TG2) as a surface-cargo of astrocytes-derived EVs. Incubation of hippocampal neurons with primed astrocyte-derived EVs have led to an increase in [Ca2+]i, unlike EVs from TG2-knockout astrocytes. Exposure of neurons or brain slices to extracellular TG2 promoted a [Ca2+]i rise, which was reversible upon TG2 removal and was dependent on Ca2+ influx through the plasma membrane. Patch-clamp and calcium imaging recordings revealed TG2-dependent neuronal membrane depolarization and activation of inward currents, due to the Na+/Ca2+-exchanger (NCX) operating in the reverse mode and indirect activation of L-type VOCCs, as indicated by VOCCs/NCX pharmacological inhibitors. A subunit of Na+/K+-ATPase was selected by comparative proteomics and identified as being functionally inhibited by extracellular TG2, implicating Na+/K+-ATPase inhibition in NCX reverse mode-switching leading to Ca2+ influx and higher basal [Ca2+]i. These data suggest that reactive astrocytes control intraneuronal [Ca2+]i through release of EVs with TG2 as responsible cargo, which could have a significant impact on synaptic activity in brain inflammation.
Collapse
Affiliation(s)
- Elisa Tonoli
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| | - Ivan Verduci
- Department of Bioscience, University of Milan, Milano 20133, Italy
| | | | - Ilaria Prada
- CNR Institute of Neuroscience, Vedano al Lambro 20854, Italy
| | - Greta Forcaia
- Human Physiology Lab., School of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
| | - Clare Coveney
- School of Science and Technology, The John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| | - Maria Pia Savoca
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| | - David J Boocock
- School of Science and Technology, The John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| | - Giulio Sancini
- Human Physiology Lab., School of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy; NeuroMI (Milan Center for Neuroscience), School of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
| | - Michele Mazzanti
- Department of Bioscience, University of Milan, Milano 20133, Italy
| | - Claudia Verderio
- CNR Institute of Neuroscience, Vedano al Lambro 20854, Italy; NeuroMI (Milan Center for Neuroscience), School of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy.
| | - Elisabetta A M Verderio
- School of Science and Technology, Centre for Health, Ageing and Understanding of Disease, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom; Biological Sciences Department (BiGeA), University of Bologna, Bologna 40126, Italy.
| |
Collapse
|
16
|
Badia-Soteras A, de Vries J, Dykstra W, Broersen LM, Verkuyl JM, Smit AB, Verheijen MHG. High-Throughput Analysis of Astrocyte Cultures Shows Prevention of Reactive Astrogliosis by the Multi-Nutrient Combination Fortasyn Connect. Cells 2022; 11:cells11091428. [PMID: 35563732 PMCID: PMC9099974 DOI: 10.3390/cells11091428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 12/23/2022] Open
Abstract
Astrocytes are specialized glial cells that tile the central nervous system (CNS) and perform numerous essential functions. Astrocytes react to various forms of CNS insults by altering their morphology and molecular profile, through a process known as reactive astrogliosis. Accordingly, astrocyte reactivity is apparent in many neurodegenerative diseases, among which one is Alzheimer’s disease (AD). Recent clinical trials on early-stage AD have demonstrated that Fortasyn Connect (FC), a multi-nutrient combination providing specific precursors and cofactors for phospholipid synthesis, helps to maintain neuronal functional connectivity and cognitive performance of patients. Several studies have shown that FC may act through its effects on neuronal survival and synaptogenesis, leading to reduced astrocyte reactivity, but whether FC can directly counteract astrocyte reactivity remains to be elucidated. Hence, we developed an in vitro model of reactive astrogliosis using the pro-inflammatory cytokines TNF-α and IFN-γ together with an automated high-throughput assay (AstroScan) to quantify molecular and morphological changes that accompany reactive astrogliosis. Next, we showed that FC is potent in preventing cytokine-induced reactive astrogliosis, a finding that might be of high relevance to understand the beneficial effects of FC-based interventions in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Aina Badia-Soteras
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Janneke de Vries
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Werner Dykstra
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Laus M. Broersen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (L.M.B.); (J.M.V.)
| | - Jan Martin Verkuyl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (L.M.B.); (J.M.V.)
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Mark H. G. Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
- Correspondence:
| |
Collapse
|
17
|
Tsai CM, Chang SF, Li CC, Chang H. Transcranial photobiomodulation (808 nm) attenuates pentylenetetrazole-induced seizures by suppressing hippocampal neuroinflammation, astrogliosis, and microgliosis in peripubertal rats. NEUROPHOTONICS 2022; 9:015006. [PMID: 35345494 PMCID: PMC8955735 DOI: 10.1117/1.nph.9.1.015006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
Significance: Transcranial photobiomodulation (tPBM) at 808 nm attenuates pentylenetetrazole (PTZ)-induced seizures and convulsive status epilepticus (CSE) in peripubertal rats by protecting neurons from injury and parvalbumin-positive interneurons from apoptosis, and preserving the integrity of perisomatic inhibitory networks. However, the effects of tPBM on neuroinflammation, astrogliosis, and microgliosis in epileptic rat brains are unknown. Thus, further study to unveil these aspects is needed for understanding the phenomena of tPBM on pediatric CSE prevention. Aim: To evaluate the effects of tPBM on neuroinflammation, astrogliosis, and microgliosis in peripubertal rat hippocampus with PTZ-induced seizures and SE. Approach: An 808-nm diode laser was applied transcranially to peripubertal rats prior to PTZ injection. Immunofluorescence staining of neuron-specific enolase (NSE) was used as a marker of neuroinflammation, glial fibrillary acid protein (GFAP) for astrogliosis, ionized calcium-binding adapter molecule 1 (Iba-1) for microgliosis, and mitochondrial cytochrome c oxidase subunit 1 (MT-CO1) for confirming the involvement of cytochrome c oxidase (CCO). Results: tPBM significantly reduced NSE immunoreactivity in CA3 in PTZ-treated rats, GFAP immunoreactivity in CA1, and Iba-1 immunoreactivity in CA3. Enhancement of hippocampal MT-CO1 reflected that tPBM acted in CCO-dependent manner. Conclusions: tPBM (808) attenuated PTZ-induced seizures and SE by suppressing neuroinflammation, astrogliosis, and microgliosis in peripubertal rats.
Collapse
Affiliation(s)
- Chung-Min Tsai
- Taipei Medical University, Graduate Institute of Medical Sciences, College of Medicine, Taipei, Taiwan
- MacKay Children’s Hospital, Department of Pediatrics, Taipei, Taiwan
| | - Shwu-Fen Chang
- Taipei Medical University, Graduate Institute of Medical Sciences, College of Medicine, Taipei, Taiwan
| | - Chih-Chuan Li
- Taipei Medical University Hospital, Department of Pediatrics, Taipei, Taiwan
| | - Hsi Chang
- Taipei Medical University Hospital, Department of Pediatrics, Taipei, Taiwan
- Taipei Medical University, College of Medicine, School of Medicine, Department of Pediatrics, Taipei, Taiwan
| |
Collapse
|
18
|
Frintrop L, Trinh S, Seitz J, Kipp M. The Role of Glial Cells in Regulating Feeding Behavior: Potential Relevance to Anorexia Nervosa. J Clin Med 2021; 11:jcm11010186. [PMID: 35011927 PMCID: PMC8745326 DOI: 10.3390/jcm11010186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/17/2021] [Accepted: 12/25/2021] [Indexed: 12/16/2022] Open
Abstract
Eating behavior is controlled by hypothalamic circuits in which agouti-related peptide-expressing neurons when activated in the arcuate nucleus, promote food intake while pro-opiomelanocortin-producing neurons promote satiety. The respective neurotransmitters signal to other parts of the hypothalamus such as the paraventricular nucleus as well as several extra-hypothalamic brain regions to orchestrate eating behavior. This complex process of food intake may be influenced by glia cells, in particular astrocytes and microglia. Recent studies showed that GFAP+ astrocyte cell density is reduced in the central nervous system of an experimental anorexia nervosa model. Anorexia nervosa is an eating disorder that causes, among the well-known somatic symptoms, brain volume loss which was associated with neuropsychological deficits while the underlying pathophysiology is unknown. In this review article, we summarize the findings of glia cells in anorexia nervosa animal models and try to deduce which role glia cells might play in the pathophysiology of eating disorders, including anorexia nervosa. A better understanding of glia cell function in the regulation of food intake and eating behavior might lead to the identification of new drug targets.
Collapse
Affiliation(s)
- Linda Frintrop
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany;
- Correspondence: ; Tel.: +49-(0)-381-494-8406
| | - Stefanie Trinh
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany;
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, 52074 Aachen, Germany;
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany;
| |
Collapse
|
19
|
Babaee A, Vaghefi SHE, Dehghani Soltani S, Asadi Shekaari M, Shahrokhi N, Basiri M. Hippocampal Astrocyte Response to Melatonin Following Neural Damage Induction in Rats. Basic Clin Neurosci 2021; 12:177-186. [PMID: 34925714 PMCID: PMC8672670 DOI: 10.32598/bcn.12.2.986.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/13/2019] [Accepted: 05/18/2020] [Indexed: 01/06/2023] Open
Abstract
Introduction: Brain injury induces an almost immediate response from glial cells, especially astrocytes. Activation of astrocytes leads to the production of inflammatory cytokines and reactive oxygen species that may result in secondary neuronal damage. Melatonin is an anti-inflammatory and antioxidant agent, and it has been reported to exert neuroprotection through the prevention of neuronal death in several models of central nervous system injury. This study aimed to investigate the effect of melatonin on astrocyte activation induced by Traumatic Brain Injury (TBI) in rat hippocampus and dentate gyrus. Methods: Animals were randomly divided into 5 groups; Sham group, TBI group, vehicle group, and melatonin-treated TBI groups (TBI+Mel5, TBI+Mel20). Immunohistochemical method (GFAP marker) and TUNEL assay were used to evaluate astrocyte reactivity and neuronal death, respectively. Results: The results demonstrated that the astrocyte number was reduced significantly in melatonin-treated groups compared to the vehicle group. Additionally, based on TUNEL results, melatonin administration noticeably reduced the number of apoptotic neurons in the rat hippocampus and dentate gyrus. Conclusion: In general, our findings suggest that melatonin treatment after brain injury reduces astrocyte reactivity as well as neuronal cell apoptosis in rat hippocampus and dentate gyrus.
Collapse
Affiliation(s)
- Abdolreza Babaee
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Samereh Dehghani Soltani
- Department of Anatomy, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Majid Asadi Shekaari
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Nader Shahrokhi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohsen Basiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
20
|
Ahn YJ, Shin HJ, Jeong EA, An HS, Lee JY, Jang HM, Kim KE, Lee J, Shin MC, Roh GS. Exendin-4 Pretreatment Attenuates Kainic Acid-Induced Hippocampal Neuronal Death. Cells 2021; 10:cells10102527. [PMID: 34685508 PMCID: PMC8534217 DOI: 10.3390/cells10102527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/15/2022] Open
Abstract
Exendin-4 (Ex-4) is a glucagon-like peptide-1 receptor (GLP-1R) agonist that protects against brain injury. However, little is known about the effect of Ex-4 on kainic acid (KA)-induced seizures and hippocampal cell death. Therefore, this study evaluated the neuroprotective effects of Ex-4 pretreatment in a mouse model of KA-induced seizures. Three days before KA treatment, mice were intraperitoneally injected with Ex-4. We found that Ex-4 pretreatment reversed KA-induced reduction of GLP-1R expression in the hippocampus and attenuated KA-induced seizure score, hippocampal neuronal death, and neuroinflammation. Ex-4 pretreatment also dramatically reduced hippocampal lipocalin-2 protein in KA-treated mice. Furthermore, immunohistochemical studies showed that Ex-4 pretreatment significantly alleviated blood–brain barrier leakage. Finally, Ex-4 pretreatment stimulated hippocampal expression of phosphorylated cyclic adenosine monophosphate (cAMP) response element-binding protein (p-CREB), a known target of GLP-1/GLP-1R signaling. These findings indicate that Ex-4 pretreatment may protect against KA-induced neuronal damage by regulating GLP-1R/CREB-mediated signaling pathways.
Collapse
Affiliation(s)
- Yu-Jeong Ahn
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Hyun-Joo Shin
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Eun-Ae Jeong
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Hyeong-Seok An
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Jong-Youl Lee
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Hye-Min Jang
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Kyung-Eun Kim
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Jaewoong Lee
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Meong-Cheol Shin
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Gyeongsang National University, Jinju 52828, Korea;
| | - Gu-Seob Roh
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
- Correspondence: ; Tel.: +82-55-772-8035; Fax: +82-55-772-8039
| |
Collapse
|
21
|
Ramezani F, Ghasemi-Kasman M, Nosratiyan N, Ghasemi S, Feizi F. Acute administration of sulfur-doped g-C3N4 induces cognitive deficits and exacerbates the levels of glial activation in mouse hippocampus. Brain Res Bull 2021; 176:54-66. [PMID: 34419511 DOI: 10.1016/j.brainresbull.2021.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/29/2021] [Accepted: 08/15/2021] [Indexed: 11/27/2022]
Abstract
During the last decades, graphitic carbon nitride (g-C3N4) has attracted increasing attention in several biomedical fields. In this study, the effects of sulfur-doped g-C3N4 (TCN) on cognitive function and histopathology of hippocampus were investigated in mice. The characteristics of synthetized sample were evaluated by X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FT-IR), Raman spectroscopy, transmission electron microscopy (TEM), field emission scanning electron microscopy (FESEM), and energy dispersive X-ray (EDX). Twenty-four male NMRI mice received vehicle, TCN at doses of 50, 150, or 500 mg/kg via gavage for one week. Morris water maze test was done to assess the cognitive function at day 14 post TCN administration. Nissl staining was used to determine the number of dark cells in the hippocampus. Immunostaining against NeuN, GFAP, and Iba1 was done to evaluate the neuronal density and levels of glial activation, respectively. Behavioral tests indicated that TCN reduces the spatial learning and memory in a dose-dependent manner. Histological evaluations showed an increased level of neuronal loss and glial activation in the hippocampus of TCN treated mice at doses of 150 and 500 mg/kg. Overall, our data indicate that TCN induces the cognitive impairment that is partly mediated via its exacerbating impacts on neuronal loss and glial activation.
Collapse
Affiliation(s)
- Farangis Ramezani
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Nasrin Nosratiyan
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Shahram Ghasemi
- Faculty of Chemistry, University of Mazandaran, Babolsar, Iran
| | - Farideh Feizi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
22
|
Xu D, Kong T, Shao Z, Liu M, Zhang R, Zhang S, Kong Q, Chen J, Cheng B, Wang C. Orexin-A alleviates astrocytic apoptosis and inflammation via inhibiting OX1R-mediated NF-κB and MAPK signaling pathways in cerebral ischemia/reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166230. [PMID: 34358627 DOI: 10.1016/j.bbadis.2021.166230] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/11/2021] [Accepted: 07/21/2021] [Indexed: 12/22/2022]
Abstract
Orexin-A (OXA) is a neuropeptide with neuroprotective effect by reducing cerebral ischemia/reperfusion injury (CIRI). Inflammation and apoptosis mediated by astrocyte activation are the key pathological mechanisms for CIRI. We thus attempted to confirm neuroprotective effects of OXA on astrocytic inflammation and apoptosis in CIRI and clarify the relative mechanisms. A middle cerebral artery occlusion and reperfusion (MCAO/R) rat model and U251 glioma cells model subjected to oxygen glucose deprivation and reperfusion (OGD/R) were established, with or without OXA treatment. Neurological deficit score was determined, and cerebral infarct volume was evaluated by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Western Blot was used to detect the expressions of NF-κB p65, p-p65, p-ERK, p-p38, GFAP, OX1R, IL-1β, TNF-α, IL-6, iNOS, Bcl-2, Bax, CytC, cleaved caspase-9 and cleaved caspase-3 in vivo and in vitro. Pro-inflammatory cytokines in cell supernatant IL-1β, TNF-α and IL-6 were determined by ELISA. Hoechst 33342 staining was used to detect the apoptosis of astrocyte. Immunofluorescent staining was performed to assess the nuclear translocation of p65 and the expression of GFAP. The results showed that OXA significantly improved neurological deficit score and decreased the volume of infarct area in brain. OXA decreased inflammatory mediators, inhibited astrocyte activation and nuclear translocation of NF-κB and phosphorylation of NF-κB, MAPK/ERK and MAPK/p38. Besides, OXA suppressed apoptosis via upregulating the ratio of Bcl-2/Bax and downregulating cytochrome C, cleaved-caspase-9 and cleaved caspase-3. Overall, it was concluded that OXA exerts neuroprotective effect during CIRI through attenuating astrocytes apoptosis, astrocytes activation and pro-inflammatory cytokines production, by Inhibiting OX1R-mediated NF-κB, MAPK/ERK and MAPK/p38 signaling pathways. The progress in our study is helpful to elucidate the molecular mechanisms of OXA neuroprotection, which could lead to the development of new treatment strategies for ischemic stroke.
Collapse
Affiliation(s)
- Dandan Xu
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | | | - Ziqi Shao
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Minghui Liu
- Basic Medical Sciences, Jining Medical University, Jining 272067, China
| | - Rumin Zhang
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Shengnan Zhang
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Qingxia Kong
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining 272029, China
| | - Jing Chen
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Baohua Cheng
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China.
| | - Chunmei Wang
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China.
| |
Collapse
|
23
|
Pekna M, Pekny M. The Complement System: A Powerful Modulator and Effector of Astrocyte Function in the Healthy and Diseased Central Nervous System. Cells 2021; 10:cells10071812. [PMID: 34359981 PMCID: PMC8303424 DOI: 10.3390/cells10071812] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/08/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
The complement system, an effector arm of the innate immune system that plays a critical role in tissue inflammation, the elimination of pathogens and the clearance of dead cells and cell debris, has emerged as a regulator of many processes in the central nervous system, including neural cell genesis and migration, control of synapse number and function, and modulation of glial cell responses. Complement dysfunction has also been put forward as a major contributor to neurological disease. Astrocytes are neuroectoderm-derived glial cells that maintain water and ionic homeostasis, and control cerebral blood flow and multiple aspects of neuronal functioning. By virtue of their expression of soluble as well as membrane-bound complement proteins and receptors, astrocytes are able to both send and receive complement-related signals. Here we review the current understanding of the multiple functions of the complement system in the central nervous system as they pertain to the modulation of astrocyte activity, and how astrocytes use the complement system to affect their environment in the healthy brain and in the context of neurological disease.
Collapse
Affiliation(s)
- Marcela Pekna
- Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne 3010, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle 2308, Australia
- Correspondence: ; Tel.: +46-31-786-3581
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden;
- Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne 3010, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle 2308, Australia
| |
Collapse
|
24
|
Koyama Y. Endothelin ET B Receptor-Mediated Astrocytic Activation: Pathological Roles in Brain Disorders. Int J Mol Sci 2021; 22:ijms22094333. [PMID: 33919338 PMCID: PMC8122402 DOI: 10.3390/ijms22094333] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
In brain disorders, reactive astrocytes, which are characterized by hypertrophy of the cell body and proliferative properties, are commonly observed. As reactive astrocytes are involved in the pathogenesis of several brain disorders, the control of astrocytic function has been proposed as a therapeutic strategy, and target molecules to effectively control astrocytic functions have been investigated. The production of brain endothelin-1 (ET-1), which increases in brain disorders, is involved in the pathophysiological response of the nervous system. Endothelin B (ETB) receptors are highly expressed in reactive astrocytes and are upregulated by brain injury. Activation of astrocyte ETB receptors promotes the induction of reactive astrocytes. In addition, the production of various astrocyte-derived factors, including neurotrophic factors and vascular permeability regulators, is regulated by ETB receptors. In animal models of Alzheimer’s disease, brain ischemia, neuropathic pain, and traumatic brain injury, ETB-receptor-mediated regulation of astrocytic activation has been reported to improve brain disorders. Therefore, the astrocytic ETB receptor is expected to be a promising drug target to improve several brain disorders. This article reviews the roles of ETB receptors in astrocytic activation and discusses its possible applications in the treatment of brain disorders.
Collapse
Affiliation(s)
- Yutaka Koyama
- Laboratory of Pharmacology, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kita Higashinada, Kobe 668-8558, Japan
| |
Collapse
|
25
|
Navarro A, García M, Rodrigues AS, Garcia PV, Camarinho R, Segovia Y. Reactive astrogliosis in the dentate gyrus of mice exposed to active volcanic environments. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2021; 84:213-226. [PMID: 33283687 DOI: 10.1080/15287394.2020.1850381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Air pollution has been associated with neuroinflammatory processes and is considered a risk factor for the development of neurodegenerative diseases. Volcanic environments are considered a natural source of air pollution. However, the effects of natural source air pollution on the central nervous system (CNS) have not been reported, despite the fact that up to 10% of the world's population lives near a historically active volcano. In order to assess the response of the CNS to such exposure, our study was conducted in the island of Sao Miguel (Azores, Portugal) in two different areas: Furnas, which is volcanically active one, and compared to Rabo de Peixe, a reference site without manifestations of active volcanism using Mus musculus as a bioindicator species. To evaluate the state of the astroglial population in the dentate gyrus in both samples, the number of astrocytes was determined using immunofluorescence methods (anti-GFAP and anti-GS). In addition, the astrocytic branches in that hippocampal area were examined. Our results showed an increase in GFAP+ astrocytes and a reduction in GS+ astrocytes in Furnas-exposed mice compared to animals from Rabo de Peixe. In addition, astrocytes in the dentate gyrus of chronically exposed animals exhibited longer branches compared to those residing at the reference site. Thus, reactive astrogliosis and astrocyte dysfunction are found in mice living in an active volcanic environment.
Collapse
Affiliation(s)
- A Navarro
- Department of Biotechnology, University of Alicante , Alicante, Spain
| | - M García
- Department of Biotechnology, University of Alicante , Alicante, Spain
| | - A S Rodrigues
- Faculty of Sciences and Technology, University of the Azores , Ponta Delgada, Portugal
- IVAR, Research Institute for Volcanology and Risk Assessment, University of the Azores , Ponta Delgada, Portugal
| | - P V Garcia
- Faculty of Sciences and Technology, University of the Azores , Ponta Delgada, Portugal
- cE3c, Centre for Ecology, Evolution and Environmental Changes, and Azorean Biodiversity Group, University of the Azores , Ponta Delgada, Portugal
| | - R Camarinho
- Faculty of Sciences and Technology, University of the Azores , Ponta Delgada, Portugal
- IVAR, Research Institute for Volcanology and Risk Assessment, University of the Azores , Ponta Delgada, Portugal
| | - Y Segovia
- Department of Biotechnology, University of Alicante , Alicante, Spain
| |
Collapse
|
26
|
Vigen TR, Brudek T, Pakkenberg B, Olesen MV. Quantitative Cellular Changes in the Thalamus of Patients with Multiple System Atrophy. Neuroscience 2021; 459:142-152. [PMID: 33577952 DOI: 10.1016/j.neuroscience.2021.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 01/10/2023]
Abstract
The thalamus is a brain region consisting of anatomical and functional connections between various spinal, subcortical, and cortical regions, which has a putative role in the clinical manifestation of Multiple System Atrophy (MSA). Previous stereological studies have reported significant anatomical alterations in diverse brain regions of MSA patients, including the cerebral cortex, basal ganglia and white matter, but no quantitative studies have examined the thalamus. To establish the extent of thalamic involvement, we applied stereological methods to estimate the total number of neurons and glial cells (oligodendrocytes, astrocytes and microglia) as well as the volume in two thalamic sub-regions, the mediodorsal nucleus (MDT) and the anterior principal nucleus (APn), in brains from ten MSA patients and 11 healthy control subjects. Compared to healthy controls, MSA patients had significantly fewer neurons (26%) in the MDT, but not the APn. We also found significantly more astrocytes (32%) and microglia (54%) in the MDT, with no such changes in the APn. Finally, we saw no group differences in the total number of oligodendrocytes. Our findings show a region-specific loss of thalamic neurons that occurs without loss of oligodendrocytes, whereas thalamic microgliosis seems to occur alongside astrogliosis. These pathological changes in the thalamus may contribute to the cognitive impairment seen in most patients with MSA.
Collapse
Affiliation(s)
- Tanya R Vigen
- Research Laboratory for Stereology and Neuroscience, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark
| | - Tomasz Brudek
- Research Laboratory for Stereology and Neuroscience, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark
| | - Bente Pakkenberg
- Research Laboratory for Stereology and Neuroscience, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark; Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| | - Mikkel V Olesen
- Research Laboratory for Stereology and Neuroscience, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Denmark
| |
Collapse
|
27
|
Leptin enhances adult neurogenesis and reduces pathological features in a transgenic mouse model of Alzheimer's disease. Neurobiol Dis 2020; 148:105219. [PMID: 33301880 DOI: 10.1016/j.nbd.2020.105219] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/18/2020] [Accepted: 12/03/2020] [Indexed: 01/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common dementia worldwide and is characterized by the presence of senile plaques by amyloid-beta (Aβ) and neurofibrillary tangles of hyperphosphorylated Tau protein. These changes lead to progressive neuronal degeneration and dysfunction, resulting in severe brain atrophy and cognitive deficits. With the discovery that neurogenesis persists in the adult mammalian brain, including brain regions affected by AD, studies of the use of neural stem cells (NSCs) for the treatment of neurodegenerative diseases to repair or prevent neuronal cell loss have increased. Here we demonstrate that leptin administration increases the neurogenic process in the dentate gyrus of the hippocampus as well as in the subventricular zone of lateral ventricles of adult and aged mice. Chronic treatment with leptin increased NSCs proliferation with significant effects on proliferation and differentiation of newborn cells. The expression of the long form of the leptin receptor, LepRb, was detected in the neurogenic niches by reverse qPCR and immunohistochemistry. Moreover, leptin modulated astrogliosis, microglial cell number and the formation of senile plaques. Additionally, leptin led to attenuation of Aβ-induced neurodegeneration and superoxide anion production as revealed by Fluoro-Jade B and dihydroethidium staining. Our study contributes to the understanding of the effects of leptin in the brain that may lead to the development of new therapies to treat Alzheimer's disease.
Collapse
|
28
|
Cutuli D, Landolfo E, Nobili A, De Bartolo P, Sacchetti S, Chirico D, Marini F, Pieroni L, Ronci M, D'Amelio M, D'Amato FR, Farioli-Vecchioli S, Petrosini L. Behavioral, neuromorphological, and neurobiochemical effects induced by omega-3 fatty acids following basal forebrain cholinergic depletion in aged mice. ALZHEIMERS RESEARCH & THERAPY 2020; 12:150. [PMID: 33198763 PMCID: PMC7667851 DOI: 10.1186/s13195-020-00705-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022]
Abstract
Background In recent years, mechanistic, epidemiologic, and interventional studies have indicated beneficial effects of omega-3 polyunsaturated fatty acids (n-3 PUFA) against brain aging and age-related cognitive decline, with the most consistent effects against Alzheimer’s disease (AD) confined especially in the early or prodromal stages of the pathology. In the present study, we investigated the action of n-3 PUFA supplementation on behavioral performances and hippocampal neurogenesis, volume, and astrogliosis in aged mice subjected to a selective depletion of basal forebrain cholinergic neurons. Such a lesion represents a valuable model to mimic one of the most reliable hallmarks of early AD neuropathology. Methods Aged mice first underwent mu-p75-saporin immunotoxin intraventricular lesions to obtain a massive cholinergic depletion and then were orally supplemented with n-3 PUFA or olive oil (as isocaloric control) for 8 weeks. Four weeks after the beginning of the dietary supplementation, anxiety levels as well as mnesic, social, and depressive-like behaviors were evaluated. Subsequently, hippocampal morphological and biochemical analyses and n-3 PUFA brain quantification were carried out. Results The n-3 PUFA treatment regulated the anxiety alterations and reverted the novelty recognition memory impairment induced by the cholinergic depletion in aged mice. Moreover, n-3 PUFA preserved hippocampal volume, enhanced neurogenesis in the dentate gyrus, and reduced astrogliosis in the hippocampus. Brain levels of n-3 PUFA were positively related to mnesic abilities. Conclusions The demonstration that n-3 PUFA are able to counteract behavioral deficits and hippocampal neurodegeneration in cholinergically depleted aged mice promotes their use as a low-cost, safe nutraceutical tool to improve life quality at old age, even in the presence of first stages of AD.
Collapse
Affiliation(s)
- Debora Cutuli
- IRCCS Fondazione Santa Lucia, Rome, Italy. .,University of Rome "Sapienza", Rome, Italy.
| | - Eugenia Landolfo
- IRCCS Fondazione Santa Lucia, Rome, Italy.,University of Rome "Sapienza", Rome, Italy
| | - Annalisa Nobili
- IRCCS Fondazione Santa Lucia, Rome, Italy.,University "Campus Bio-Medico", Rome, Italy
| | - Paola De Bartolo
- IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Human Sciences, Guglielmo Marconi University, Rome, Italy
| | | | - Doriana Chirico
- Institute of Biochemistry and Cell Biology, CNR, Monterotondo, Italy
| | - Federica Marini
- Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | | | - Maurizio Ronci
- Department of Pharmacy, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Marcello D'Amelio
- IRCCS Fondazione Santa Lucia, Rome, Italy.,University "Campus Bio-Medico", Rome, Italy
| | | | | | | |
Collapse
|
29
|
Huang J, Liu Y, Cheng L, Li J, Zhang T, Zhao G, Zhang H. Glucagon-like peptide-1 cleavage product GLP-1(9-36) reduces neuroinflammation from stroke via the activation of insulin-like growth factor 1 receptor in astrocytes. Eur J Pharmacol 2020; 887:173581. [PMID: 32949596 DOI: 10.1016/j.ejphar.2020.173581] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is an endogenous gut hormone and a key regulator in maintaining glucose homeostasis by stimulating insulin secretion. Its natural cleavage product GLP-1 (9-36), which was formerly considered a "bio-inactive" metabolite mainly due to its low affinity for GLP-1 receptor, possesses unique properties such as cardiovascular protection. Little is known about the effects and mechanisms of GLP-1 (9-36) in cerebral ischemia and reperfusion injury. Here, we report that systemic application of GLP-1 (9-36) in adult mice facilitated functional recovery and reduced infarct volume, astrogliosis, and neuronal apoptosis following middle cerebral artery occlusion and reperfusion. Interestingly, these effects were still observed in GLP-1 receptor knockout (Glp-1rKO) mice but were partially reversed in insulin-like growth factor 1 (IGF-1) receptor knockdown (Igf-1rKD) mice. Primary astrocytes were cultured and subjected to oxygen-glucose deprivation/reoxygenation (OGD/R), and enzyme-linked immunosorbent assay indicated that GLP-1 (9-36) pretreatment reduces tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 levels. This effect was not diminished in Glp-1rKO astrocytes but was reversed in Igf-1rKO astrocytes, emphasizing that the anti-inflammatory effect of GLP-1 (9-36) in astrocytes is independent of GLP-1 receptor signaling and is instead mediated by IGF-1 receptor. Immunoprecipitation experiments showed that GLP-1 (9-36) directly interacts with IGF-1 receptor in astrocytes. Western blot data indicated that GLP-1 (9-36) activates IGF-1 receptor and downstream PI3K-AKT pathway in astrocytes upon OGD/R injury, which was abrogated by preincubation with IGF-1 receptor autophosphorylation inhibitor picropodophyllin. Thus, our findings suggest that GLP-1 (9-36) improved stroke outcome by reducing inflammation in astrocytes via interaction with IGF-1 receptor.
Collapse
Affiliation(s)
- Jing Huang
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China; Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yunhan Liu
- Department of Neurology Impatient, Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Liusiyuan Cheng
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China
| | - Jihong Li
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China
| | - Tangrui Zhang
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Huinan Zhang
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, China.
| |
Collapse
|
30
|
Huang Y, Liao Y, Zhang H, Li S. Lead exposure induces cell autophagy via blocking the Akt/mTOR signaling in rat astrocytes. J Toxicol Sci 2020; 45:559-567. [PMID: 32879255 DOI: 10.2131/jts.45.559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Lead is a main threat to human health due to its neurotoxicity and the astrocyte is known to be a common deposit site of lead in vivo. However, the detailed mechanisms related to lead exposure in the astrocytes were unclear. In order to deeply investigate this issue, we used Sprague-Dawley (SD) rats and astrocytes isolated from the hippocampus of SD rats to establish the lead-exposed animal and cell models through treating with lead acetate. The expression levels of GFAP, LC3, and p62 in the rat hippocampus were detected by immunofluorescence and Western blot after lead exposure. The effects of autophagy on lead-exposed astrocytes were studied by further autophagy inhibitor 3-methyladenine (3-MA) induction. Transmission electron microscopy was used to observe autophagosomes in astrocytes after lead acetate treatment, followed by assessing related autophagy protein markers. In addition, some inflammatory cytokines and oxidative stress markers were also evaluated after lead exposure and 3-MA administration. We found that lead exposure induced activation of astrocytes, as evidenced by increased GFAP levels and GFAP-positive staining cells in the rat hippocampus. Moreover, lead exposure induced autophagy in astrocytes, as evidenced by increased LC3II and Beclin 1 protein levels and decreased p62 expression in both the rat hippocampus and astrocytes, and it was confirmed that this autophagy was activated through blocking the downstream Akt/target of the rapamycin (mTOR) pathway in astrocytes. Furthermore, it was shown that treatment of lead acetate increased the release of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), and the accumulation of malondialdehyde (MDA) and myeloperoxidase (MPO) in astrocytes, which could be alleviated by further 3-MA induction. Therefore, we conclude that lead exposure can induce the autophagy of astrocytes via blocking the Akt/mTOR pathway, leading to accelerated release of inflammatory factors and oxidative stress indicators in astrocytes.
Collapse
Affiliation(s)
- Yingying Huang
- Department of Physiology, School of Life Sciences, China Medical University, China.,School of Nursing, Jinzhou Medical University, China
| | - Yingjun Liao
- Department of Physiology, School of Life Sciences, China Medical University, China
| | - Huijun Zhang
- School of Nursing, Jinzhou Medical University, China
| | - Shuyun Li
- School of Nursing, Jinzhou Medical University, China
| |
Collapse
|
31
|
Extracellular Alpha-Synuclein Promotes a Neuroinhibitory Secretory Phenotype in Astrocytes. Life (Basel) 2020; 10:life10090183. [PMID: 32911644 PMCID: PMC7555668 DOI: 10.3390/life10090183] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 11/16/2022] Open
Abstract
Multiple system atrophy (MSA) and dementia with Lewy bodies (DLB) are α-synucleinopathies that exhibit widespread astrogliosis as a component of the neuroinflammatory response. Munc18, a protein critical to vesicle exocytosis, was previously found to strongly mark morphologically activated astrocytes in brain tissue of MSA patients. Immunofluorescence of MSA, DLB and normal brain tissue sections was combined with cell culture and co-culture experiments to investigate the relationship between extracellular α-synuclein and the transition to a secretory astrocyte phenotype. Increased Munc18-positive vesicles were resolved in activated astrocytes in MSA and DLB tissue compared to controls, and they were also significantly upregulated in the human 1321N1 astrocytoma cell line upon treatment with α-synuclein, with parallel increases in GFAP expression and IL-6 secretion. In co-culture experiments, rat primary astrocytes pretreated with α-synuclein inhibited the growth of neurites of co-cultured primary rat neurons and upregulated chondroitin sulphate proteoglycan. Taken together, these results indicate that the secretory machinery is significantly upregulated in the astrocyte response to extracellular α-synuclein and may participate in the release of neuroinhibitory and proinflammatory factors in α-synucleinopathies.
Collapse
|
32
|
Werkman IL, Lentferink DH, Baron W. Macroglial diversity: white and grey areas and relevance to remyelination. Cell Mol Life Sci 2020; 78:143-171. [PMID: 32648004 PMCID: PMC7867526 DOI: 10.1007/s00018-020-03586-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023]
Abstract
Macroglia, comprising astrocytes and oligodendroglial lineage cells, have long been regarded as uniform cell types of the central nervous system (CNS). Although regional morphological differences between these cell types were initially described after their identification a century ago, these differences were largely ignored. Recently, accumulating evidence suggests that macroglial cells form distinct populations throughout the CNS, based on both functional and morphological features. Moreover, with the use of refined techniques including single-cell and single-nucleus RNA sequencing, additional evidence is emerging for regional macroglial heterogeneity at the transcriptional level. In parallel, several studies revealed the existence of regional differences in remyelination capacity between CNS grey and white matter areas, both in experimental models for successful remyelination as well as in the chronic demyelinating disease multiple sclerosis (MS). In this review, we provide an overview of the diversity in oligodendroglial lineage cells and astrocytes from the grey and white matter, as well as their interplay in health and upon demyelination and successful remyelination. In addition, we discuss the implications of regional macroglial diversity for remyelination in light of its failure in MS. Since the etiology of MS remains unknown and only disease-modifying treatments altering the immune response are available for MS, the elucidation of macroglial diversity in grey and white matter and its putative contribution to the observed difference in remyelination efficiency between these regions may open therapeutic avenues aimed at enhancing endogenous remyelination in either area.
Collapse
Affiliation(s)
- Inge L Werkman
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA
| | - Dennis H Lentferink
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Wia Baron
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, the Netherlands.
| |
Collapse
|
33
|
Astrocyte and Oligodendrocyte Cross-Talk in the Central Nervous System. Cells 2020; 9:cells9030600. [PMID: 32138223 PMCID: PMC7140446 DOI: 10.3390/cells9030600] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/23/2022] Open
Abstract
Over the last decade knowledge of the role of astrocytes in central nervous system (CNS) neuroinflammatory diseases has changed dramatically. Rather than playing a merely passive role in response to damage it is clear that astrocytes actively maintain CNS homeostasis by influencing pH, ion and water balance, the plasticity of neurotransmitters and synapses, cerebral blood flow, and are important immune cells. During disease astrocytes become reactive and hypertrophic, a response that was long considered to be pathogenic. However, recent studies reveal that astrocytes also have a strong tissue regenerative role. Whilst most astrocyte research focuses on modulating neuronal function and synaptic transmission little is known about the cross-talk between astrocytes and oligodendrocytes, the myelinating cells of the CNS. This communication occurs via direct cell-cell contact as well as via secreted cytokines, chemokines, exosomes, and signalling molecules. Additionally, this cross-talk is important for glial development, triggering disease onset and progression, as well as stimulating regeneration and repair. Its critical role in homeostasis is most evident when this communication fails. Here, we review emerging evidence of astrocyte-oligodendrocyte communication in health and disease. Understanding the pathways involved in this cross-talk will reveal important insights into the pathogenesis and treatment of CNS diseases.
Collapse
|
34
|
Lee EJ, Han JC, Park DY, Kee C. A neuroglia-based interpretation of glaucomatous neuroretinal rim thinning in the optic nerve head. Prog Retin Eye Res 2020; 77:100840. [PMID: 31982595 DOI: 10.1016/j.preteyeres.2020.100840] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/02/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022]
Abstract
Neuroretinal rim thinning (NRR) is a characteristic glaucomatous optic disc change. However, the precise mechanism of the rim thinning has not been completely elucidated. This review focuses on the structural role of the glioarchitecture in the formation of the glaucomatous NRR thinning. The NRR is a glia-framed structure, with honeycomb geometry and mechanically reinforced astrocyte processes along the transverse plane. When neural damage selectively involves the neuron and spares the glia, the gross structure of the tissue is preserved. The disorganization and loss of the glioarchitecture are the two hallmarks of optic nerve head (ONH) remodeling in glaucoma that leads to the thinning of NRR tissue upon axonal loss. This is in contrast to most non-glaucomatous optic neuropathies with optic disc pallor where hypertrophy of the glioarchitecture is associated with the seemingly absent optic disc cupping. Arteritic anterior ischemic optic neuropathy is an exception where pan-necrosis of ONH tissue leads to NRR thinning. Milder ischemia indicates selective neuronal loss that spares glia in non-arteritic anterior ischemic optic neuropathy. The biological reason is the heterogeneous glial response determined by the site, type, and severity of the injury. The neuroglial interpretation explains how the cellular changes underlie the clinical findings. Updated understandings on glial responses illustrate the mechanical, microenvironmental, and microglial modulation of activated astrocytes in glaucoma. Findings relevant to the possible mechanism of the astrocyte death in advanced glaucoma are also emerging. Ultimately, a better understanding of glaucomatous glial response may lead to glia-targeting neuroprotection in the future.
Collapse
Affiliation(s)
- Eun Jung Lee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Jong Chul Han
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Do Young Park
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Changwon Kee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea.
| |
Collapse
|
35
|
Udovin LD, Kobiec T, Herrera MI, Toro-Urrego N, Kusnier CF, Kölliker-Frers RA, Ramos-Hryb AB, Luaces JP, Otero-Losada M, Capani F. Partial Reversal of Striatal Damage by Palmitoylethanolamide Administration Following Perinatal Asphyxia. Front Neurosci 2020; 13:1345. [PMID: 31969800 PMCID: PMC6960201 DOI: 10.3389/fnins.2019.01345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 11/29/2019] [Indexed: 01/27/2023] Open
Abstract
Perinatal asphyxia (PA) is a clinical condition brought by a birth temporary oxygen deprivation associated with long-term damage in the corpus striatum, one of the most compromised brain areas. Palmitoylethanolamide (PEA) is a neuromodulator well known for its protective effects in brain injury models, including PA, albeit not deeply studied regarding its particular effects in the corpus striatum following PA. Using Bjelke et al. (1991) PA model, full-term pregnant rats were decapitated, and uterus horns were placed in a water bath at 37°C for 19 min. One hour later, the pups were injected with PEA 10 mg/kg s.c., and placed with surrogate mothers. After 30 days, the animals were perfused, and coronal striatal sections were collected to analyze protein-level expression by Western blot and the reactive area by immunohistochemistry for neuron markers: phosphorylated neurofilament-heavy/medium-chain (pNF-H/M) and microtubule-associated protein-2 (MAP-2), and the astrocyte marker, glial fibrillary acidic protein (GFAP). Results indicated that PA produced neuronal damage and morphological changes. Asphyctic rats showed a decrease in pNF-H/M and MAP-2 reactive areas, GFAP+ cells number, and MAP-2 as well as pNF-H/M protein expression in the striatum. Treatment with PEA largely restored the number of GFAP+ cells. Most important, it ameliorated the decrease in pNF-H/M and MAP-2 reactive areas in asphyctic rats. Noticeably, PEA treatment reversed the decrease in MAP-2 protein expression and largely prevented PA-induced decrease in pNF-H/M protein expression. PA did not affect the GFAP protein level. Treatment with PEA attenuated striatal damage induced by PA, suggesting its therapeutic potential for the prevention of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lucas D Udovin
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Tamara Kobiec
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina.,Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - María I Herrera
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina.,Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Nicolás Toro-Urrego
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Carlos F Kusnier
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Rodolfo A Kölliker-Frers
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Ana B Ramos-Hryb
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Juan P Luaces
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina
| | - Francisco Capani
- Institute of Cardiological Research, University of Buenos Aires, National Research Council (ININCA-UBA-CONICET), Buenos Aires, Argentina.,Departamento de Biología, Universidad Argentina John F. Kennedy (UAJK), Buenos Aires, Argentina
| |
Collapse
|
36
|
Thangaraj A, Sil S, Tripathi A, Chivero ET, Periyasamy P, Buch S. Targeting endoplasmic reticulum stress and autophagy as therapeutic approaches for neurological diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 350:285-325. [PMID: 32138902 DOI: 10.1016/bs.ircmb.2019.11.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
37
|
Diviccaro S, Melcangi RC, Giatti S. Post-finasteride syndrome: An emerging clinical problem. Neurobiol Stress 2019; 12:100209. [PMID: 32435662 PMCID: PMC7231981 DOI: 10.1016/j.ynstr.2019.100209] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022] Open
Abstract
The presence of side effects during pharmacological treatment is unfortunately a quite common problem. In this review, we focused our attention on adverse events related to 5 alpha-reductase (5α-R) inhibitors (i.e., finasteride and dutasteride), approved for the treatment of benign prostatic hyperplasia and androgenetic alopecia (AGA). Although these drugs are generally well tolerated, many reports described adverse effects in men during treatment, such as sexual dysfunction and mood alteration. In addition, it has been also reported that persistent side effects may occur in some AGA patients. This condition, termed post-finasteride syndrome (PFS) is characterized by sexual side effects (i.e., low libido, erectile dysfunction, decreased arousal and difficulty in achieving orgasm), depression, anxiety and cognitive complaints that are still present despite drug withdrawal. Indeed, some national agencies (e.g., Swedish Medical Products Agency, the Medicines and Healthcare Products Regulatory Agency of UK and the U.S. Food and Drug Administration) required to include multiple persistent side effects within the finasteride labels. As here reported, these observations are mainly based on self-reporting of the symptomatology by the patients and few clinical studies have been performed so far. In addition, molecular mechanisms and/or genetic determinants behind such adverse effects have been poorly explored both in patients and animal models. Therefore, results here discussed indicate that PFS is an emerging clinical problem that needs to be further elucidated.
Collapse
Affiliation(s)
- Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| | - Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| |
Collapse
|
38
|
Liu H, Wu X, Luo J, Wang X, Guo H, Feng D, Zhao L, Bai H, Song M, Liu X, Guo W, Li X, Yue L, Wang B, Qu Y. Pterostilbene Attenuates Astrocytic Inflammation and Neuronal Oxidative Injury After Ischemia-Reperfusion by Inhibiting NF-κB Phosphorylation. Front Immunol 2019; 10:2408. [PMID: 31681297 PMCID: PMC6811521 DOI: 10.3389/fimmu.2019.02408] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022] Open
Abstract
Astrocyte-mediated inflammation and oxidative stress elicit cerebral ischemia-reperfusion (IR) injury after stroke. Nuclear factor (NF)-κB activates astrocytes and generates pro-inflammatory factors. The purpose of the present study is to elucidate the effect of pterostilbene (PTE, a natural stilbene) on astrocytic inflammation and neuronal oxidative injury following cerebral ischemia-reperfusion injury. A middle cerebral artery occlusion-reperfusion (MCAO/R) mouse model and HT22/U251 co-culture model subjected to oxygen-glucose deprivation and re-introduction (OGD/R) were employed, with or without PTE treatment. The data showed that PTE delivery immediately after reperfusion, at 1 h after occlusion, decreased infarct volume, brain edema, and neuronal apoptosis and improved long-term neurological function. PTE decreased oxidation (i.e., production of reactive oxygen species, malondialdehyde) and inflammatory mediators (tumor necrosis factor-α, interleukin-1β, and interleukin-6) and increased anti-oxidative enzyme activities (i.e., of superoxide dismutase, glutathione peroxidase), by inhibiting phosphorylation and nuclear translocation of NF-κB. In conclusion, PTE attenuated astrocyte-mediated inflammation and oxidative injury following IR via NF-κB inhibition. Overall, PTE is a promising neuroprotective agent.
Collapse
Affiliation(s)
- Haixiao Liu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xun Wu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jianing Luo
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaogang Wang
- Department of Neurosurgery, The 960th Hospital, Jinan, China
| | - Hao Guo
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lei Zhao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hao Bai
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Mingyang Song
- Department of Nursing, The 960th Hospital, Jinan, China
| | - Xunyuan Liu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Guo
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xia Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Liang Yue
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bodong Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China.,Department of Neurosurgery, The 960th Hospital, Jinan, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
39
|
Espitia Pinzon N, van Mierlo H, de Jonge JC, Brevé JJP, Bol JGJM, Drukarch B, van Dam AM, Baron W. Tissue Transglutaminase Promotes Early Differentiation of Oligodendrocyte Progenitor Cells. Front Cell Neurosci 2019; 13:281. [PMID: 31312122 PMCID: PMC6614186 DOI: 10.3389/fncel.2019.00281] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/11/2019] [Indexed: 01/09/2023] Open
Abstract
Demyelinated lesions of the central nervous system are characteristic for multiple sclerosis (MS). Remyelination is not very effective, particular at later stages of the disease, which results in a chronic neurodegenerative character with worsening of symptoms. Previously, we have shown that the enzyme Tissue Transglutaminase (TG2) is downregulated upon differentiation of oligodendrocyte progenitor cells (OPCs) into myelin-forming oligodendrocytes and that TG2 knock-out mice lag behind in remyelination after cuprizone-induced demyelination. Here, we examined whether astrocytic or oligodendroglial TG2 affects OPCs in a cell-specific manner to modulate their differentiation, and therefore myelination. Our findings indicate that human TG2-expressing astrocytes did not modulate OPC differentiation and myelination. In contrast, persistent TG2 expression upon OPC maturation or exogenously added recombinant TG2 accelerated OPC differentiation and myelin membrane formation. Continuous exposure of recombinant TG2 to OPCs at different consecutive developmental stages, however, decreased OPC differentiation and myelin membrane formation, while it enhanced myelination in dorsal root ganglion neuron-OPC co-cultures. In MS lesions, TG2 is absent in OPCs, while human OPCs show TG2 immunoreactivity during brain development. Exposure to the MS-relevant pro-inflammatory cytokine IFN-γ increased TG2 expression in OPCs and prolonged expression of endogenous TG2 upon differentiation. However, despite the increased TG2 levels, OPC maturation was not accelerated, indicating that TG2-mediated OPC differentiation may be counteracted by other pathways. Together, our data show that TG2, either endogenously expressed, or exogenously supplied to OPCs, accelerates early OPC differentiation. A better understanding of the role of TG2 in the OPC differentiation process during MS is of therapeutic interest to overcome remyelination failure.
Collapse
Affiliation(s)
- Nathaly Espitia Pinzon
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Hanneke van Mierlo
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jenny C de Jonge
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - John J P Brevé
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - John G J M Bol
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Benjamin Drukarch
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Anne-Marie van Dam
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Wia Baron
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
40
|
Zhang Y, Bao Y, Qiu W, Peng L, Fang L, Xu Y, Yang H. Structural and visual functional deficits in a rat model of neuromyelitis optica spectrum disorders related optic neuritis. Exp Eye Res 2018; 175:124-132. [DOI: 10.1016/j.exer.2018.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/30/2018] [Accepted: 06/13/2018] [Indexed: 12/15/2022]
|
41
|
Ikeshima-Kataoka H, Matsui Y, Uede T. Osteopontin is indispensable for activation of astrocytes in injured mouse brain and primary culture. Neurol Res 2018; 40:1071-1079. [PMID: 30246619 DOI: 10.1080/01616412.2018.1517995] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
OBJECTIVES Osteopontin (OPN) is an inflammatory cytokine inducer involved in cell proliferation and migration in inflammatory diseases or tumors. To investigate the function of OPN in astrocyte activation during brain injury, we compared OPN-deficient (OPN/KO) with wild-type (WT) mouse brains after stab wound injury and primary culture of astrocytes. METHODS Primary cultures of astrocytes were prepared from either WT or OPN/KO postnatal mouse brains. Activation efficiency of astrocytes in primary culture was accessed using Western blotting by examining the protein levels of glial fibrillary acidic protein (GFAP) and tenascin-C (TN-C), which are markers for reactive astrocytes, following lipopolysaccharide (LPS) stimulation. Furthermore, the stab wound injury on the cerebral cortex as a brain traumatic injury model was used, and activation of astrocytes and microglial cells was investigated using immunofluorescent analysis on fixed brain sections. RESULTS Primary cultures of astrocytes prepared from WT or OPN/KO postnatal mouse brains showed that only 25% of normal shaped astrocytes in a flask were produced in OPN/KO mice. The expression levels of both GFAP and TN-C were downregulated in the primary culture of astrocytes from OPN/KO mice compared with that from WT mice. By the immunofluorescent analysis on the injured brain sections, glial activation was attenuated in OPN/KO mice compared with WT mice. DISCUSSION Our data suggest that OPN is essential for proper astrocytic generation in vitro culture prepared from mouse cerebral cortex. OPN is indispensable for astrocyte activation in the mouse brain injury model and in LPS stimulated primary culture. ABBREVIATIONS AQP4: aquaporin 4; BBB: blood brain barrier; BrdU: bromo-deoxy uridine; CNS: central nervous system; GFAP: glial fibllirary acidic protein; IgG: immunoglobulin G; LPS: lipopolysaccharide; OPN: osteopontin; OPN/KO: osteopontin-deficient; TN-C: tenascin-C.
Collapse
Affiliation(s)
- Hiroko Ikeshima-Kataoka
- a Department of Pharmacology and Neuroscience , Keio University School of Medicine , Tokyo , Japan.,b Faculty of Science and Engineering , Waseda University , Tokyo , Japan
| | - Yutaka Matsui
- c Department of Cardiovascular Medicine , Tonan Hospital , Sapporo , Hokkaido , Japan.,d Department of Matrix Medicine, Institute of Genetic Medicine , Hokkaido University , Sapporo , Hokkaido , Japan
| | - Toshimitsu Uede
- d Department of Matrix Medicine, Institute of Genetic Medicine , Hokkaido University , Sapporo , Hokkaido , Japan
| |
Collapse
|
42
|
Pekny M, Wilhelmsson U, Tatlisumak T, Pekna M. Astrocyte activation and reactive gliosis-A new target in stroke? Neurosci Lett 2018; 689:45-55. [PMID: 30025833 DOI: 10.1016/j.neulet.2018.07.021] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/03/2018] [Accepted: 07/14/2018] [Indexed: 11/27/2022]
Abstract
Stroke is an acute insult to the central nervous system (CNS) that triggers a sequence of responses in the acute, subacute as well as later stages, with prominent involvement of astrocytes. Astrocyte activation and reactive gliosis in the acute stage of stroke limit the tissue damage and contribute to the restoration of homeostasis. Astrocytes also control many aspects of neural plasticity that is the basis for functional recovery. Here, we discuss the concept of intermediate filaments (nanofilaments) and the complement system as two handles on the astrocyte responses to injury that both present attractive opportunities for novel treatment strategies modulating astrocyte functions and reactive gliosis.
Collapse
Affiliation(s)
- Milos Pekny
- Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 440, 40530 Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; University of Newcastle, Newcastle, NSW, Australia.
| | - Ulrika Wilhelmsson
- Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 440, 40530 Gothenburg, Sweden
| | - Turgut Tatlisumak
- Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 440, 40530 Gothenburg, Sweden; Department of Neurology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marcela Pekna
- Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 440, 40530 Gothenburg, Sweden; Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
43
|
Liu CY, Yang Y, Ju WN, Wang X, Zhang HL. Emerging Roles of Astrocytes in Neuro-Vascular Unit and the Tripartite Synapse With Emphasis on Reactive Gliosis in the Context of Alzheimer's Disease. Front Cell Neurosci 2018; 12:193. [PMID: 30042661 PMCID: PMC6048287 DOI: 10.3389/fncel.2018.00193] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 06/14/2018] [Indexed: 01/09/2023] Open
Abstract
Astrocytes, which are five-fold more numerous than neurons in the central nervous system (CNS), are traditionally viewed to provide simple structural and nutritional supports for neurons and to participate in the composition of the blood brain barrier (BBB). In recent years, the active roles of astrocytes in regulating cerebral blood flow (CBF) and in maintaining the homeostasis of the tripartite synapse have attracted increasing attention. More importantly, astrocytes have been associated with the pathogenesis of Alzheimer's disease (AD), a major cause of dementia in the elderly. Although microglia-induced inflammation is considered important in the development and progression of AD, inflammation attributable to astrogliosis may also play crucial roles. A1 reactive astrocytes induced by inflammatory stimuli might be harmful by up-regulating several classical complement cascade genes thereby leading to chronic inflammation, while A2 induced by ischemia might be protective by up-regulating several neurotrophic factors. Here we provide a concise review of the emerging roles of astrocytes in the homeostasis maintenance of the neuro-vascular unit (NVU) and the tripartite synapse with emphasis on reactive astrogliosis in the context of AD, so as to pave the way for further research in this area, and to search for potential therapeutic targets of AD.
Collapse
Affiliation(s)
- Cai-Yun Liu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Yang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Wei-Na Ju
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hong-Liang Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
- Department of Life Sciences, The National Natural Science Foundation of China, Beijing, China
| |
Collapse
|
44
|
Morphine-Mediated Brain Region-Specific Astrocytosis Involves the ER Stress-Autophagy Axis. Mol Neurobiol 2018; 55:6713-6733. [PMID: 29344928 DOI: 10.1007/s12035-018-0878-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/07/2018] [Indexed: 01/08/2023]
Abstract
A recent study from our lab has revealed a link between morphine-mediated autophagy and synaptic impairment. The current study was aimed at investigating whether morphine-mediated activation of astrocytes involved the ER stress/autophagy axis. Our in vitro findings demonstrated upregulation of GFAP indicating astrocyte activation with a concomitant increase in the production of proinflammatory cytokines in morphine-exposed human astrocytes. Using both pharmacological and gene-silencing approaches, it was demonstrated that morphine-mediated defective autophagy involved upstream activation of ER stress with subsequent downstream astrocyte activation via the μ-opioid receptor (MOR). In vivo validation demonstrated preferential activation of ER stress/autophagy axis in the areas of the brain not associated with pain such as the basal ganglia, frontal cortex, occipital cortex, and the cerebellum of morphine-dependent rhesus macaques, and this correlated with increased astrocyte activation and neuroinflammation. Interventions aimed at blocking either the MOR or ER stress could thus likely be developed as promising therapeutic targets for abrogating morphine-mediated astrocytosis.
Collapse
|
45
|
Anti-encephalitogenic effects of ethyl pyruvate are reflected in the central nervous system and the gut. Biomed Pharmacother 2017; 96:78-85. [DOI: 10.1016/j.biopha.2017.09.110] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/20/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022] Open
|
46
|
Ferrer I. Diversity of astroglial responses across human neurodegenerative disorders and brain aging. Brain Pathol 2017; 27:645-674. [PMID: 28804999 PMCID: PMC8029391 DOI: 10.1111/bpa.12538] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022] Open
Abstract
Astrogliopathy refers to alterations of astrocytes occurring in diseases of the nervous system, and it implies the involvement of astrocytes as key elements in the pathogenesis and pathology of diseases and injuries of the central nervous system. Reactive astrocytosis refers to the response of astrocytes to different insults to the nervous system, whereas astrocytopathy indicates hypertrophy, atrophy/degeneration and loss of function and pathological remodeling occurring as a primary cause of a disease or as a factor contributing to the development and progression of a particular disease. Reactive astrocytosis secondary to neuron loss and astrocytopathy due to intrinsic alterations of astrocytes occur in neurodegenerative diseases, overlap each other, and, together with astrocyte senescence, contribute to disease-specific astrogliopathy in aging and neurodegenerative diseases with abnormal protein aggregates in old age. In addition to the well-known increase in glial fibrillary acidic protein and other proteins in reactive astrocytes, astrocytopathy is evidenced by deposition of abnormal proteins such as β-amyloid, hyper-phosphorylated tau, abnormal α-synuclein, mutated huntingtin, phosphorylated TDP-43 and mutated SOD1, and PrPres , in Alzheimer's disease, tauopathies, Lewy body diseases, Huntington's disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease, respectively. Astrocytopathy in these diseases can also be manifested by impaired glutamate transport; abnormal metabolism and release of neurotransmitters; altered potassium, calcium and water channels resulting in abnormal ion and water homeostasis; abnormal glucose metabolism; abnormal lipid and, particularly, cholesterol metabolism; increased oxidative damage and altered oxidative stress responses; increased production of cytokines and mediators of the inflammatory response; altered expression of connexins with deterioration of cell-to-cell networks and transfer of gliotransmitters; and worsening function of the blood brain barrier, among others. Increased knowledge of these aspects will permit a better understanding of brain aging and neurodegenerative diseases in old age as complex disorders in which neurons are not the only players.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- Institute of NeuropathologyPathologic Anatomy Service, Bellvitge University Hospital, IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos IIIMadridSpain
| |
Collapse
|
47
|
Regulation of neuroinflammation by matrix metalloproteinase-8 inhibitor derivatives in activated microglia and astrocytes. Oncotarget 2017; 8:78677-78690. [PMID: 29108257 PMCID: PMC5667990 DOI: 10.18632/oncotarget.20207] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 07/25/2017] [Indexed: 11/25/2022] Open
Abstract
Matrix metalloproteinases (MMPs) play a pivotal role in neuroinflammation that is associated with neurodegenerative diseases. Our group recently reported that MMP-8 mediates inflammatory reactions by modulating the processing of TNF-α. To improve the efficacy of the currently available MMP-8 inhibitor (M8I), we have synthesized structurally modified M8I derivatives (comp 2, 3, 4, 5) and compared their efficacy with original compound (comp 1). Among M8I derivatives, comp 2, 3, and 5 inhibited the production of NO, ROS, and IL-6 more efficiently than the original compound in lipopolysaccharide (LPS)-stimulated microglia. When we compared the anti-inflammatory mechanisms of the most effective derivative, comp 3, with comp 1, comp 3 suppressed the mRNA expression of iNOS and cytokines more efficiently than comp 1. Although comp 1 inhibits only TNF-α processing, comp 3 additionally inhibits the expression of TNF-α. Both compounds inhibited LPS-induced activity of MAP kinases, NF-κB, and AP-1, while they increased heme oxygenase-1 expression by upregulating AMPK-Nrf2 signaling. Overall, the effect of comp 3 on anti-inflammatory signaling was much stronger than comp 1. We verified the anti-inflammatory effects of comp 1 and 3 in the LPS-injected mouse brain and primary cultured astrocytes. Comp 1 and 3 suppressed microglial activation, astrogliosis, and proinflammatory gene expression in the brain. Moreover, the compounds inhibited proinflammatory gene expression in the cultured astrocytes. Collectively, our data suggest that the MMP-8 inhibitor may be a promising therapeutic agent for neuroinflammatory disorders.
Collapse
|
48
|
Williams PA, Marsh-Armstrong N, Howell GR. Neuroinflammation in glaucoma: A new opportunity. Exp Eye Res 2017; 157:20-27. [PMID: 28242160 PMCID: PMC5497582 DOI: 10.1016/j.exer.2017.02.014] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 02/16/2017] [Accepted: 02/23/2017] [Indexed: 10/20/2022]
Abstract
Mounting evidence suggests neuroinflammation is a key process in glaucoma, yet the precise roles are not known. Understanding these complex processes, which may also be a key in other common neurodegenerations such as Alzheimer's disease, will lead to targeted therapeutics for a disease that affects as many as 80 million people worldwide. Here, we define neuroinflammation as any immune-relevant response by a variety of cell types including astrocytes, microglia, and peripherally derived cells occurring in the optic nerve head and/or retina. In this review article, we first discuss clinical evidence for neuroinflammation in glaucoma and define neuroinflammation in glaucoma. We then review the inflammatory pathways that have been associated with glaucoma. Finally, we set out key research directions that we believe will greatly advance our understanding of the role of neuroinflammation in glaucoma. This review arose from a discussion of neuroinflammation in glaucoma at the 2015 meeting of The Lasker/IRRF Initiative for Innovation in Vision Science. This manuscript sets out to summarize one of these sessions; "Inflammation and Glaucomatous Neurodegeneration", as well as to review the current state of the literature surrounding neuroinflammation in glaucoma.
Collapse
Affiliation(s)
| | - Nick Marsh-Armstrong
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, USA; Graduate Program of Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|
49
|
Espitia Pinzon N, Sanz-Morello B, Brevé JJP, Bol JGJM, Drukarch B, Bauer J, Baron W, van Dam AM. Astrocyte-derived tissue Transglutaminase affects fibronectin deposition, but not aggregation, during cuprizone-induced demyelination. Sci Rep 2017; 7:40995. [PMID: 28128219 PMCID: PMC5269585 DOI: 10.1038/srep40995] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/13/2016] [Indexed: 02/07/2023] Open
Abstract
Astrogliosis as seen in Multiple Sclerosis (MS) develops into astroglial scarring, which is beneficial because it seals off the site of central nervous system (CNS) damage. However, astroglial scarring also forms an obstacle that inhibits axon outgrowth and (re)myelination in brain lesions. This is possibly an important cause for incomplete remyelination in the CNS of early stage MS patients and for failure in remyelination when the disease progresses. In this study we address whether under demyelinating conditions in vivo, tissue Transglutaminase (TG2), a Ca2+ -dependent enzyme that catalyses posttranslational modification of proteins, contributes to extracellular matrix (ECM) deposition and/or aggregation. We used the cuprizone model for de- and remyelination. TG2 immunoreactivity and enzymatic activity time-dependently appeared in astrocytes and ECM, respectively, in the corpus callosum of cuprizone-treated mice. Enhanced presence of soluble monomeric and multimeric fibronectin was detected during demyelination, and fibronectin immunoreactivity was slightly decreased in cuprizone-treated TG2-/- mice. In vitro TG2 overexpression in astrocytes coincided with more, while knock-down of TG2 with less fibronectin production. TG2 contributes, at least partly, to fibronectin production, and may play a role in fibronectin deposition during cuprizone-induced demyelination. Our observations are of interest in understanding the functional implications of TG2 during astrogliosis.
Collapse
Affiliation(s)
- Nathaly Espitia Pinzon
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, 1081 HV, The Netherlands
| | - Berta Sanz-Morello
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, 1081 HV, The Netherlands
| | - John J. P. Brevé
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, 1081 HV, The Netherlands
| | - John G. J. M. Bol
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, 1081 HV, The Netherlands
| | - Benjamin Drukarch
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, 1081 HV, The Netherlands
| | - Jan Bauer
- Center for Brain Research, Dept. Neuroimmunology, Vienna, A-1090, Austria
| | - Wia Baron
- University Medical Center Groningen, Dept. of Cell Biology, Groningen, 9713 AV, The Netherlands
| | - Anne-Marie van Dam
- VU University Medical Center, Neuroscience Campus Amsterdam, Dept. Anatomy and Neurosciences, Amsterdam, 1081 HV, The Netherlands
| |
Collapse
|
50
|
Saavedra LM, Fenton Navarro B, Torner L. Early Life Stress Activates Glial Cells in the Hippocampus but Attenuates Cytokine Secretion in Response to an Immune Challenge in Rat Pups. Neuroimmunomodulation 2017; 24:242-255. [PMID: 29332092 DOI: 10.1159/000485383] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/04/2017] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Early life stress (ELS) increases the vulnerability to developing psychopathological disorders in adulthood that are accompanied by brain inflammatory processes. However, it is not known how a combined double hit (stress and immune) at an early age affects the response of the neuroimmune system. Here we investigated the effect of periodic maternal separation (MS) followed by administration of lipopolysaccharide (LPS) on glial cells in the CA3 region and hilus of the hippocampus and on cytokine release on postnatal day (PN) 15. METHODS Male rat pups were subjected to MS (3 h/day, PN1-14). MS and control pups received a single LPS injection (1 mg/kg of body weight) on PN14. They were subjected to an open field test 1 h later. The pups were sacrificed 90 min after LPS injection (PN14) or on PN15 for cytokine or immunohistological analyses, respectively. RESULTS LPS reduced the locomotion and induced high corticosterone levels in treated pups. MS or LPS reduced microglial density and activated microglial cells in the hippocampal CA3 and hilus regions. Microglial activation was highest in MS-LPS pups. The astrocyte density was mildly reduced by MS or LPS in the CA3 region and hilus, but the reduction was maximal in MS-LPS pups. LPS increased the secretion of plasmatic interleukin (IL)-1β, tumor necrosis factor-α, and IL-6, and of hippocampal IL-1β protein, but these were attenuated in MS-LPS pups. CONCLUSION Although MS and LPS activate neuroimmune cells, stress attenuates the hippocampal and peripheral cytokine response to LPS through an as-yet unidentified adaptive mechanism. These results provide information regarding the neurobiology of stress and inflammation.
Collapse
Affiliation(s)
- Luis Miguel Saavedra
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | | | | |
Collapse
|