1
|
Hammad AM, Syaj H, Abusara OH, Khdair SI, Debas R, Hall FS. Anxiety-like behavior in rats during periods of abstinence following E-cigarette vapor and cigarette smoke exposure: Role of inflammatory cytokines and glutamate receptors. Behav Brain Res 2025; 488:115600. [PMID: 40287018 DOI: 10.1016/j.bbr.2025.115600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Prolonged exposure to cigarette smoke induces changes in the glutamatergic systems as well as neuroinflammation. We examined E-cigarette vapor and cigarette smoke effects on inflammatory cytokines and metabotropic glutamate receptors. Furthermore, we investigated the behavioral changes related to E-cigarette vapor and cigarette smoke exposure through utilizing open field (OF), elevated plus maze (EPM) and light/dark (LD) tests. Male Sprague-Dawley rats were randomly assigned to three experimental groups: Control, E-cigarette, and Cigarettes groups. Exposure to either E-cigarette vapor or cigarette smoke exposure was performed for 2 hr/day, 5 days/week, for 60 days. Behavioral tests were conducted every two weeks, 24 hr after exposure, during periods of abstinence. Anxiety-like behaviors were increased following repeated periods of abstinence from E-cigarette vapor or cigarette smoke. E-cigarette vapor and cigarette smoke elevated the relative mRNA expression of nuclear factor ĸB (Nf-ĸB), interleukin 6 (Il-6), and metabolic glutamate receptor 5 (mglur5) and reduced expression of interleukin 1β (Il-1β), tumor necrosis α (Tnf-α), and metabolic glutamate receptor 2 (mglur2) in prefrontal cortex (PFC) and nucleus accumbens (NAc). Moreover, no effect was observed on nuclear factor erythroid 2 (Nrf2), metabolic glutamate receptor 1 (mglur1), or metabolic glutamate receptor 3 (mglur3) expression. E-cigarette vapor and cigarette smoke exposure can lead to abstinence-induced anxiety-like behavior partially through molecular changes in the PFC and NAc.
Collapse
Affiliation(s)
- Alaa M Hammad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan.
| | - Heba Syaj
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Osama H Abusara
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Sawsan I Khdair
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Rasha Debas
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
2
|
Denver P, Cunningham C. Microglial activation and neuroinflammation in acute and chronic cognitive deficits in sepsis. Neuropharmacology 2025; 267:110285. [PMID: 39746541 DOI: 10.1016/j.neuropharm.2024.110285] [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: 09/20/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Sepsis is characterised by dysregulated immune responses to infection, leading to multi-organ dysfunction and high rates of mortality. With increasing survival rates in recent years long-term neurological and psychiatric consequences have become more apparent in survivors. Many patients develop sepsis associated encephalopathy (SAE) which encompasses the profound but usually transient neuropsychiatric syndrome delirium but also new brain injury that emerges in the months and years post-sepsis. It is now clear that systemic inflammatory signals reach the brain during sepsis and that very significant neuroinflammation ensues. The major brain resident immune cell population, the microglia, has been implicated in acute and chronic cognitive dysfunction in animal models of sepsis based on a growing number of studies using bacterial endotoxin and in polymicrobial sepsis models such as cecal ligation and puncture. The current review explores the effects of sepsis on the brain, focussing on how systemic insults translate to microglial activation and neuroinflammation and how this disrupts neuronal function and integrity. We examine what has been demonstrated specifically with respect to microglial activation, revealing robust evidence for a role for neuroinflammation in sepsis-induced brain sequelae but less clear information on the extent of the specific microglial contribution to this, arising from findings using global knockout mice, non-selective drugs and treatments that equally target peripheral and central compartments. There is, nonetheless, clear evidence that microglia do become activated and do contribute to brain consequences of sepsis thus arguing for improved understanding of these neuroinflammatory processes toward the prevention and treatment of sepsis-induced brain dysfunction.
Collapse
Affiliation(s)
- Paul Denver
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland.
| |
Collapse
|
3
|
Cai Y, Yu F, Wu W, Chen W. Study on the mechanism of Dexmedetomidine's effect on postoperative cognitive dysfunction in elderly people. Front Physiol 2025; 16:1508661. [PMID: 40144543 PMCID: PMC11936804 DOI: 10.3389/fphys.2025.1508661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication among elderly patients following surgical procedures, significantly impairing postoperative recovery and quality of life. The selection and dosage of intraoperative anaesthetic drugs are frequently implicated as contributing factors in the development of POCD. In recent years, dexmedetomidine (DEX), a novel α2-adrenoceptor agonist, has been increasingly utilized in surgical anaesthesia for elderly patients, showing potential as both a preventive and therapeutic agent for POCD. This paper provides a comprehensive review of current research on the mechanisms by which DEX affects POCD in the elderly. Additionally, it explores DEX's mechanisms of action in the context of neuroprotection, anti-inflammation, antioxidative stress, and the regulation of apoptosis, autophagy, and analgesia. The objective is to provide reliable theoretical support and a reference point for the clinical application of DEX in POCD among the elderly, thereby promoting its broader use in clinical practice to improve outcomes and enhance quality of life.
Collapse
Affiliation(s)
- Yuanbin Cai
- Department of Anesthesiology, Putuo District Central Hospital, Shanghai, China
| | - Fan Yu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Wei Wu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Wurong Chen
- Department of Anesthesiology, Putuo District Central Hospital, Shanghai, China
| |
Collapse
|
4
|
El-Naggar AE, Helmy MM, El-Gowilly SM, El-Mas MM. The Cholinergic Amelioration of Sepsis-Induced Baroreflex Dysfunction and Brainstem Inflammation Is Negated by Central Adenosine A3 Receptors. Pharmaceuticals (Basel) 2025; 18:388. [PMID: 40143165 PMCID: PMC11946792 DOI: 10.3390/ph18030388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Sepsis has been shown to depress arterial baroreceptor function, and this effect is counterbalanced by the cholinergic anti-inflammatory pathway. Considering the importance of central adenosine receptors in baroreceptor function, this study tested whether central adenosine A3 receptors (A3ARs) modulate the cholinergic-baroreflex interaction in sepsis and whether this interaction is modulated by mitogen-activated protein kinases (MAPKs) and related proinflammatory cytokines. Methods: Sepsis was induced by cecal ligation and puncture (CLP) and rats were instrumented with femoral and intracisternal (i.c.) catheters. Baroreflex sensitivity (BRS) was measured 24 h later in conscious animals using the vasoactive method, which correlates changes in blood pressure caused by i.v. phenylephrine (PE) and sodium nitroprusside (SNP) to concomitant reciprocal changes in heart rate. Results: The reduction in reflex bradycardic (BRS-PE), but not tachycardic (BRS-SNP), responses elicited by CLP was reversed by i.v. nicotine in a dose-related manner. The BRS-PE effect of nicotine was blunted following intracisternal administration of IB-MECA (A3AR agonist, 4 µg/rat). The depressant action of IB-MECA on the BRS facilitatory action of nicotine was abrogated following central inhibition of MAPK-JNK (SP 600125), PI3K (wortmannin), and TNFα (infliximab), but not MAPK-ERK (PD 98059). Additionally, the nicotine suppression of sepsis-induced upregulation of NFκB and NOX2 expression in the nucleus tractus solitarius (NTS) was negated by A3AR activation. The molecular effect of IB-MECA on NFκB expression disappeared in the presence of SP 600125, wortmannin, or infliximab. Conclusions: The central PI3K/MAPK-JNK/TNFα pathway contributes to the restraining action of A3ARs on cholinergic amelioration of sepsis-induced central neuroinflammatory responses and impairment of the baroreceptor-mediated negative chronotropism.
Collapse
Affiliation(s)
- Amany E. El-Naggar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21511, Egypt; (A.E.E.-N.); (M.M.H.); (S.M.E.-G.)
| | - Mai M. Helmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21511, Egypt; (A.E.E.-N.); (M.M.H.); (S.M.E.-G.)
| | - Sahar M. El-Gowilly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21511, Egypt; (A.E.E.-N.); (M.M.H.); (S.M.E.-G.)
| | - Mahmoud M. El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21511, Egypt; (A.E.E.-N.); (M.M.H.); (S.M.E.-G.)
- Department of Pharmacology and Toxicology, Faculty of Medicine, College of Medicine, Kuwait University, Jabriya 46301, Kuwait
| |
Collapse
|
5
|
Li C, Ren H, Liu H, Li T, Liu Y, Wu B, Han K, Zang S, Zhao G, Wang X. Middle frontal gyrus volume mediates the relationship between interleukin-1β and antidepressant response in major depressive disorder. J Affect Disord 2025; 372:56-65. [PMID: 39592061 DOI: 10.1016/j.jad.2024.11.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024]
Abstract
Inflammation is a leading biological risk factor contributing to unfavorable outcomes of major depressive disorder (MDD). Both inflammation and depression are associated with similar alterations in brain structure, indicating that brain structural alterations could serve as a mediating factor in the adverse influence of inflammation on clinical outcomes in MDD. Nonetheless, longitudinal research has yet to confirm this hypothesis. Therefore, this study aimed at elucidating the relationships between peripheral inflammatory cytokines, gray matter volume (GMV) alterations, and antidepressant response in MDD. We studied 104 MDD patients treated with selective serotonin reuptake inhibitors and 85 healthy controls (HCs). Antidepressant response was assessed after 8-week antidepressant treatment by changes in 17-item Hamilton Depression Rating Scale (HAMD-17) scores. The GMV alterations were investigated using a voxel-based morphometry analysis. Inflammatory cytokines were measured using flow cytometry. Partial correlations were used to explore the relationships between inflammatory cytokines, GMV alterations, and antidepressant response. Compared to HCs, MDD patients showed reduced GMVs primarily in the frontal-limbic area, right insula, and right superior temporal gyrus. Furthermore, the alterations in GMVs, particularly in the right middle frontal gyrus and the left anterior cingulate gyrus, were associated with ΔHAMD-17 and inflammatory cytokines. Additionally, GMV alterations in the right middle frontal gyrus mediated the negative relationship between interleukin -1β and ΔHAMD-17. This study contributes to understanding the effect of inflammation on the brain and their relationships with antidepressant response, offering a potential explanation for the connection between inflammatory status and treatment efficacy.
Collapse
Affiliation(s)
- Cuicui Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Honghong Ren
- Department of Psychology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hongzhu Liu
- School of Medical Imaging, Binzhou Medical University, Yantai, Shandong, China
| | - Tong Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yigang Liu
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Baolin Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Ke Han
- Department of Rehabilitation, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan, Shandong, China
| | - Shuqi Zang
- Department of Rehabilitation, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan, Shandong, China
| | - Guoqing Zhao
- Department of Psychology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
6
|
Mulvaney SW, Dineen KJ, Mahadevan S, Desronvilles R, Rae Olmsted KL. Three-Month Durability of Bilateral Two-Level Stellate Ganglion Blocks in Patients with Generalized Anxiety Disorder: A Retrospective Analysis. Brain Sci 2025; 15:188. [PMID: 40002521 PMCID: PMC11853295 DOI: 10.3390/brainsci15020188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Purpose: Determine if performing ultrasound-guided, bilateral, two-level cervical sympathetic chain blocks (2LCSB) (performed on subsequent days) provides durable improvement in symptoms associated with anxiety. Methods: A retrospective chart review was conducted between January 2022 and November 2024. We identified 114 patients who received bilateral, 2LCSB for anxiety symptoms. Generalized Anxiety Disorder 7-Item Scale (GAD-7) outcome measure scores were collected at baseline and three-months post procedure in 71 males and 43 females. Results: Out of 114 patients, 99 patients (86.8%) showed a long-lasting improvement in their GAD-7 scores. Collected GAD-7 forms had a baseline average of 15.52 (14.99 for males and 16.40 for females), which decreased after three months to an average of 7.28 (6.96 for males and 7.81 for females). This represents a 52% average improvement in anxiety symptoms. Conclusions: In individuals treated with bilateral, 2LCSB, GAD-related symptoms were improved by 52% for at least 3 months regardless of initial anxiety severity.
Collapse
Affiliation(s)
- Sean W. Mulvaney
- Department of Military and Emergency Medicine, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA;
| | - Kyle J. Dineen
- Orthobiologics Research Initiative Inc., 11200 Rockville Pike #230, North Bethesda, MD 20852, USA; (S.M.); (R.D.J.)
| | - Sanjay Mahadevan
- Orthobiologics Research Initiative Inc., 11200 Rockville Pike #230, North Bethesda, MD 20852, USA; (S.M.); (R.D.J.)
| | - Roosevelt Desronvilles
- Orthobiologics Research Initiative Inc., 11200 Rockville Pike #230, North Bethesda, MD 20852, USA; (S.M.); (R.D.J.)
| | | |
Collapse
|
7
|
Liu S, Hao J, Yu T, Tuchin VV, Li J, Li D, Zhu D. Diabetes Mellitus Impairs Blood-Brain Barrier Integrality and Microglial Reactivity. JOURNAL OF BIOPHOTONICS 2025:e202400482. [PMID: 39870511 DOI: 10.1002/jbio.202400482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/29/2025]
Abstract
Diabetes mellitus (DM), a chronic metabolic disorder that adversely affects the blood-brain barrier (BBB) and microglial function in the central nervous system (CNS), contributing to neuronal damage and neurodegenerative diseases. However, the underlying molecular mechanisms linking diabetes to BBB dysfunction and microglial dysregulation remain poorly understood. Here, we assessed the impacts of diabetes on BBB and microglial reactivity and investigated its mechanisms. We found diabetes severely disrupted the BBB integrity and microglial response to vascular injury. We also revealed a potential relationship between BBB disruption and impaired microglial function, whereby increasing BBB permeability led to a downregulation of microglial P2RY12 expression, thereby impairing microglial protection against cerebrovascular injury. Understanding these mechanisms may contribute to the developing of therapeutic strategies for diabetes-related neurological complications.
Collapse
Affiliation(s)
- Shaojun Liu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Wuhan National Laboratory for Optoelectronics-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
| | - Jie Hao
- Affiliated Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Wuhan National Laboratory for Optoelectronics-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
| | - Valery V Tuchin
- Institute of Physics and Science Medical Center, Saratov State University, Saratov, Russian Federation
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Tomsk, Russian Federation
- Institute of Precision Mechanics and Control, FRS "Saratov Scientific Centre of the RAS", Saratov, Russian Federation
| | - Junming Li
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Dongyu Li
- Wuhan National Laboratory for Optoelectronics-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
- School of Optical Electronic Information-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics-MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Wuhan National Laboratory for Optoelectronics-Advanced Biomedical Imaging Facility, HUST, Wuhan, China
| |
Collapse
|
8
|
Strohm AO, Johnston C, Hernady E, Marples B, O'Banion MK, Majewska AK. Cranial irradiation disrupts homeostatic microglial dynamic behavior. J Neuroinflammation 2024; 21:82. [PMID: 38570852 PMCID: PMC10993621 DOI: 10.1186/s12974-024-03073-z] [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: 12/18/2023] [Accepted: 03/22/2024] [Indexed: 04/05/2024] Open
Abstract
Cranial irradiation causes cognitive deficits that are in part mediated by microglia, the resident immune cells of the brain. Microglia are highly reactive, exhibiting changes in shape and morphology depending on the function they are performing. Additionally, microglia processes make dynamic, physical contacts with different components of their environment to monitor the functional state of the brain and promote plasticity. Though evidence suggests radiation perturbs homeostatic microglia functions, it is unknown how cranial irradiation impacts the dynamic behavior of microglia over time. Here, we paired in vivo two-photon microscopy with a transgenic mouse model that labels cortical microglia to follow these cells and determine how they change over time in cranial irradiated mice and their control littermates. We show that a single dose of 10 Gy cranial irradiation disrupts homeostatic cortical microglia dynamics during a 1-month time course. We found a lasting loss of microglial cells following cranial irradiation, coupled with a modest dysregulation of microglial soma displacement at earlier timepoints. The homogeneous distribution of microglia was maintained, suggesting microglia rearrange themselves to account for cell loss and maintain territorial organization following cranial irradiation. Furthermore, we found cranial irradiation reduced microglia coverage of the parenchyma and their surveillance capacity, without overtly changing morphology. Our results demonstrate that a single dose of radiation can induce changes in microglial behavior and function that could influence neurological health. These results set the foundation for future work examining how cranial irradiation impacts complex cellular dynamics in the brain which could contribute to the manifestation of cognitive deficits.
Collapse
Affiliation(s)
- Alexandra O Strohm
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Carl Johnston
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
9
|
Khan MAS, Chang SL. Alcohol and the Brain-Gut Axis: The Involvement of Microglia and Enteric Glia in the Process of Neuro-Enteric Inflammation. Cells 2023; 12:2475. [PMID: 37887319 PMCID: PMC10605902 DOI: 10.3390/cells12202475] [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: 07/16/2023] [Revised: 09/25/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
Binge or chronic alcohol consumption causes neuroinflammation and leads to alcohol use disorder (AUD). AUD not only affects the central nervous system (CNS) but also leads to pathologies in the peripheral and enteric nervous systems (ENS). Thus, understanding the mechanism of the immune signaling to target the effector molecules in the signaling pathway is necessary to alleviate AUD. Growing evidence shows that excessive alcohol consumption can activate neuroimmune cells, including microglia, and change the status of neurotransmitters, affecting the neuroimmune system. Microglia, like peripheral macrophages, are an integral part of the immune defense and represent the reticuloendothelial system in the CNS. Microglia constantly survey the CNS to scavenge the neuronal debris. These cells also protect parenchymal cells in the brain and spinal cord by repairing nerve circuits to keep the nervous system healthy against infectious and stress-derived agents. In an activated state, they become highly dynamic and mobile and can modulate the levels of neurotransmitters in the CNS. In several ways, microglia, enteric glial cells, and macrophages are similar in terms of causing inflammation. Microglia also express most of the receptors that are constitutively present in macrophages. Several receptors on microglia respond to the inflammatory signals that arise from danger-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), endotoxins (e.g., lipopolysaccharides), and stress-causing molecules (e.g., alcohol). Therefore, this review article presents the latest findings, describing the roles of microglia and enteric glial cells in the brain and gut, respectively, and their association with neurotransmitters, neurotrophic factors, and receptors under the influence of binge and chronic alcohol use, and AUD.
Collapse
Affiliation(s)
- Mohammed A. S. Khan
- Department of Neurosurgery, Brigham Hospital for Children, Harvard Medical School, Boston, MA 02115, USA;
| | - Sulie L. Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ 07079, USA
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| |
Collapse
|
10
|
Wang Q, Sun J, Chen T, Song S, Hou Y, Feng L, Fan C, Li M. Ferroptosis, Pyroptosis, and Cuproptosis in Alzheimer's Disease. ACS Chem Neurosci 2023; 14:3564-3587. [PMID: 37703318 DOI: 10.1021/acschemneuro.3c00343] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023] Open
Abstract
Alzheimer's disease (AD), the most common type of dementia, is a neurodegenerative disorder characterized by progressive cognitive dysfunction. Epidemiological investigation has demonstrated that, after cardiovascular and cerebrovascular diseases, tumors, and other causes, AD has become a major health issue affecting elderly individuals, with its mortality rate acutely increasing each year. Regulatory cell death is the active and orderly death of genetically determined cells, which is ubiquitous in the development of living organisms and is crucial to the regulation of life homeostasis. With extensive research on regulatory cell death in AD, increasing evidence has revealed that ferroptosis, pyroptosis, and cuproptosis are closely related to the occurrence, development, and prognosis of AD. This paper will review the molecular mechanisms of ferroptosis, pyroptosis, and cuproptosis and their regulatory roles in AD to explore potential therapeutic targets for the treatment of AD.
Collapse
Affiliation(s)
- Qi Wang
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Jingyi Sun
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Tian Chen
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Siyu Song
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Yajun Hou
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Lina Feng
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Cundong Fan
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, Shandong, China
| | - Mingquan Li
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| |
Collapse
|
11
|
Uzcategui NL, Güçer S, Richter C, Speidel A, Zirdum E, Duszenko M, Garaschuk O, Figarella K. Live imaging of microglia during sleeping sickness reveals early and heterogeneous inflammatory responses. Front Immunol 2023; 14:1253648. [PMID: 37781403 PMCID: PMC10534015 DOI: 10.3389/fimmu.2023.1253648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Invasion of the central nervous system (CNS) is the most serious consequence of Trypanosoma brucei infection, which causes sleeping sickness. Recent experimental data have revealed some more insights into the disease during the meningoencephalitic stage. However, detailed cellular processes befalling the CNS during the disease are poorly understood. Methods To further address this issue, we implanted a cranial window on the cortex of B6.129P2(Cg)-Cx3cr1tm1Litt/J mice, infected them with Trypanosoma brucei expressing RFP via intraperitoneal injection, and monitored microglial cells and parasites longitudinally over 30 days using in vivo 2-photon imaging. We correlated the observed changes with histological analyses to evaluate the recruitment of peripheral immune cells. Results and discussion We uncovered an early involvement of microglia that precedes invasion of the CNS by the parasite. We accomplished a detailed characterization of the progressive sequence of events that correlates with microglial morphological changes and microgliosis. Our findings unveiled a heterogeneous microglial response in places of initial homeostatic disruption near brain barriers and pointed out an exceptional capability of microglia to hamper parasite proliferation inside the brain. We also found early signs of inflammation in the meninges, which synchronize with the microglial response. Moreover, we observed a massive infiltration of peripheral immune cells into the parenchyma as a signature in the final disease stage. Overall, our study provides new insights into the host-pathogen immune interactions in the meningeal and parenchymal compartments of the neocortex.
Collapse
Affiliation(s)
- Nestor L. Uzcategui
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
- Institute of Tropical Medicine, University of Tuebingen, Tuebingen, Germany
- Institute of Anatomy, Central University of Venezuela, Caracas, Venezuela
| | - Sena Güçer
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Cris Richter
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Annika Speidel
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Elizabeta Zirdum
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Michael Duszenko
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| | - Katherine Figarella
- Department of Neurophysiology, Institute of Physiology, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
12
|
Gigliotta A, Trontti K, Väänänen J, Hovatta I. Gene expression profiling reveals a role of immune system and inflammation in innate and stress-induced anxiety-like behavior. Front Genet 2023; 14:1173376. [PMID: 37260777 PMCID: PMC10229056 DOI: 10.3389/fgene.2023.1173376] [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: 02/24/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023] Open
Abstract
Anxiety is an evolutionarily conserved response that is essential for survival. Pathological anxiety, however, is a maladaptive response to nonthreatening situations and greatly affects quality of life. The recent COVID-19 pandemic has increased the prevalence of anxiety symptoms and highlighted the urge to identify the molecular events that initiate pathological anxiety. To this aim, we investigated the extent of similarity of brain region-specific gene expression patterns associated with innate and stress-induced anxiety-like behavior. We compared the cortico-frontal (FCx) and hippocampal (Hpc) gene expression patterns of five inbred mouse strains with high or low levels of innate anxiety-like behavior with gene expression patterns of mice subjected to chronic social defeat stress. We found significantly large overlap of the Hpc but small overlap of the FCx gene expression patterns in innate and stress-induced anxiety, that however, converged onto common inflammation and immune system canonical pathways. Comparing the gene expression data with drug-gene interaction datasets revealed drug candidates, including medrysone, simvastatin, captopril, and sulpiride, that produced gene expression changes opposite to those observed in innate or stress-induced anxiety-like behavior. Together, our data provide a comprehensive overview of FCx and Hpc gene expression differences between innate and stress-induced anxiety and support the role of inflammation and immune system in anxiety-like behavior.
Collapse
|
13
|
Jung H, Lee D, You H, Lee M, Kim H, Cheong E, Um JW. LPS induces microglial activation and GABAergic synaptic deficits in the hippocampus accompanied by prolonged cognitive impairment. Sci Rep 2023; 13:6547. [PMID: 37085584 PMCID: PMC10121592 DOI: 10.1038/s41598-023-32798-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/03/2023] [Indexed: 04/23/2023] Open
Abstract
Neuroinflammation impacts the brain and cognitive behavior through microglial activation. In this study, we determined the temporal sequence from microglial activation to synaptic dysfunction and cognitive behavior induced by neuroinflammation in mice. We found that LPS injection activated microglia within a short period, followed by impairments in GABAergic synapses, and that these events led to long-term cognitive impairment. We demonstrated that, 3 days after LPS injection, microglia in the hippocampus were significantly activated due to the LPS-induced inflammation in association with alterations in cellular morphology, microglial density, and expression of phagocytic markers. GABAergic synaptic impairments were detected at 4-6 days after LPS treatment, a time when microglia activity had returned to normal. Consequently, memory impairment persisted for 6 days after injection of LPS. Our results suggest that neuroinflammation induces microglia activation, GABAergic synaptic deficits and prolonged memory impairment over a defined temporal sequence. Our observations provide insight into the temporal sequence of neuroinflammation-associated brain pathologies. Moreover, the specific loss of inhibitory synapses accompanying the impaired inhibitory synaptic transmission provides mechanistic insight that may explain the prolonged cognitive deficit observed in patients with neuroinflammation. Thus, this study provides essential clues regarding early intervention strategies against brain pathologies accompanying neuroinflammation.
Collapse
Affiliation(s)
- Hyeji Jung
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea
| | - Dongsu Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Heejung You
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Myungha Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Hyeonho Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea.
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea.
- Center for Synapse Diversity and Specificity, DGIST, 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea.
| |
Collapse
|
14
|
Namakin K, Moghaddam MH, Sadeghzadeh S, Mehranpour M, Vakili K, Fathi M, Golshan A, Bayat AH, Tajik AH, Eskandari N, Mohammadzadeh I, Benisi SZ, Aliaghaei A, Abdollahifar MA. Elderberry diet improves gut-brain axis dysfunction, neuroinflammation, and cognitive impairment in the rat model of irritable bowel syndrome. Metab Brain Dis 2023; 38:1555-1572. [PMID: 36877342 DOI: 10.1007/s11011-023-01187-6] [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: 10/24/2022] [Accepted: 02/13/2023] [Indexed: 03/07/2023]
Abstract
Irritable bowel syndrome (IBS) is related to a problem in the gut-brain axis. This experimental research aimed to shed light on the potential therapeutic application of elderberry (EB), which can work on the axis and get better the IBS symptoms. There were three groups (36 Sprague-Dawley rats) in this experiment, including control, IBS, and IBS with EB diet (IBS + EB). Making use of intracolonic instillation of 1 ml of 4% acetic acid for 30 s, IBS was induced. 7 days later, the EB extract (2%) was added to the diets of all animals for 8 weeks. Some histological, behavioral, and stereological techniques were used to detect the effects of EB on the gut and brain tissues. The findings showed that the EB diet improved locomotion and decreased anxiety-like behavior in the rat models of IBS. Moreover, the diet dropped the expression of TNF-α and increased mucosal layer thickness and the number of goblet and mast cells in colon tissue samples. In the hippocampal samples, administration of EB prevented astrogliosis and astrocyte reactivity. Although hippocampal and cortical neurons decreased markedly in the IBS group, EB prevented the drop in the number of neurons. Although lots of research is needed to elucidate the effectiveness of EB in IBS and its exact molecular mechanism, the result of this study showed that EB as an antioxidant and immune-modulatory agent could be a promising research target to prevent the impairment in the gut-brain axis, and could ameliorative classic IBS symptoms.
Collapse
Affiliation(s)
- Kosar Namakin
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Sara Sadeghzadeh
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Mehranpour
- Department of Genetics, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Golshan
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Hossein Bayat
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir-Hossein Tajik
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Eskandari
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Ibrahim Mohammadzadeh
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, 1385/768, Tehran, Iran
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad-Amin Abdollahifar
- 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.
| |
Collapse
|
15
|
Yick LW, Ma OKF, Chan EYY, Yau KX, Kwan JSC, Chan KH. T follicular helper cells contribute to pathophysiology in a model of neuromyelitis optica spectrum disorders. JCI Insight 2023; 8:161003. [PMID: 36649074 PMCID: PMC9977492 DOI: 10.1172/jci.insight.161003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) are inflammatory autoimmune disorders of the CNS. IgG autoantibodies targeting the aquaporin-4 water channel (AQP4-IgGs) are the pathogenic effector of NMOSD. Dysregulated T follicular helper (Tfh) cells have been implicated in loss of B cell tolerance in autoimmune diseases. The contribution of Tfh cells to disease activity and therapeutic potential of targeting these cells in NMOSD remain unclear. Here, we established an autoimmune model of NMOSD by immunizing mice against AQP4 via in vivo electroporation. After AQP4 immunization, mice displayed AQP4 autoantibodies in blood circulation, blood-brain barrier disruption, and IgG infiltration in spinal cord parenchyma. Moreover, AQP4 immunization induced motor impairments and NMOSD-like pathologies, including astrocytopathy, demyelination, axonal loss, and microglia activation. These were associated with increased splenic Tfh, Th1, and Th17 cells; memory B cells; and plasma cells. Aqp4-deficient mice did not display motor impairments and NMOSD-like pathologies after AQP4 immunization. Importantly, abrogating ICOS/ICOS-L signaling using anti-ICOS-L antibody depleted Tfh cells and suppressed the response of Th1 and Th17 cells, memory B cells, and plasma cells in AQP4-immunized mice. These findings were associated with ameliorated motor impairments and spinal cord pathologies. This study suggests a role of Tfh cells in the pathophysiology of NMOSD in a mouse model with AQP4 autoimmunity and provides an animal model for investigating the immunological mechanisms underlying AQP4 autoimmunity and developing therapeutic interventions targeting autoimmune reactions in NMOSD.
Collapse
|
16
|
Hattori Y. The microglia-blood vessel interactions in the developing brain. Neurosci Res 2023; 187:58-66. [PMID: 36167249 DOI: 10.1016/j.neures.2022.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/03/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022]
Abstract
Microglia are the immune cells in the central nervous system (CNS). Once microglial progenitors are generated in the yolk sac, these cells enter the CNS and colonize its structures by migrating and proliferating during development. Although the microglial population in the CNS is still low in this stage compared to adults, these cells can associate with many surrounding cells, such as neural lineage cells and vascular-structure-composing cells, by extending their filopodia and with their broad migration capacity. Previous studies revealed multifaceted microglial actions on neural lineage cells, such as regulating the differentiation of neural progenitors and modulating neuronal positioning. Notably, microglia not only act on neural lineage cells but also interact with blood vessels, for example, by supporting vascular formation and integrity. On the other hand, blood vessels contribute to microglial colonization into the CNS and their migration at local tissues. Importantly, pericytes, the cells that encompass vascular endothelial cells, have been suggested to play a profound role in microglial function. This review summarizes recent advances in the understanding of the interaction of microglia and blood vessels, especially focusing on the significance of this interaction in CNS development, and discusses how microglial and blood vessel dysfunction leads to developmental disorders.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
17
|
Hattori Y. The Multiple Roles of Pericytes in Vascular Formation and Microglial Functions in the Brain. Life (Basel) 2022; 12:1835. [PMID: 36362989 PMCID: PMC9699346 DOI: 10.3390/life12111835] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 10/15/2023] Open
Abstract
In the capillary walls, vascular endothelial cells are covered with mural cells, such as smooth muscle cells and pericytes. Although pericytes had been thought to play simply a structural role, emerging evidence has highlighted their multiple functions in the embryonic, postnatal, and adult brain. As the central nervous system (CNS) develops, the brain's vascular structure gradually matures into a hierarchical network, which is crucial for the proper development of neural lineage cells by providing oxygen and nutrients. Pericytes play an essential role in vascular formation and regulate blood‒brain barrier (BBB) integrity as a component of the neurovascular unit (NVU), in collaboration with other cells, such as vascular endothelial cells, astrocytes, neurons, and microglia. Microglia, the resident immune cells of the CNS, colonize the brain at embryonic day (E) 9.5 in mice. These cells not only support the development and maturation of neural lineage cells but also help in vascular formation through their extensive migration. Recent studies have demonstrated that pericytes directly contact microglia in the CNS, and their interactions have a profound effect on physiological and pathological aspects. This review summarizes the function of pericytes, focusing on the interplay between pericytes and microglia.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
18
|
Franco R, Lillo A, Navarro G, Reyes-Resina I. The adenosine A 2A receptor is a therapeutic target in neurological, heart and oncogenic diseases. Expert Opin Ther Targets 2022; 26:791-800. [DOI: 10.1080/14728222.2022.2136570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Rafael Franco
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neurobiology laboratory, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain
| | - Alejandro Lillo
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neuropharmacology laboratory, Department of Biochemistry and Physiology. School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neuropharmacology laboratory, Department of Biochemistry and Physiology. School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
| | - Irene Reyes-Resina
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain
- Molecular Neuropharmacology laboratory, Department of Biochemistry and Physiology. School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Lai W, Huang J, Fang W, Deng S, Xie Y, Wang W, Qiao T, Xu G, Wang X, Ding F. Optic nerve head: A gatekeeper for vitreous infectious insults? Front Immunol 2022; 13:987771. [PMID: 36203577 PMCID: PMC9531234 DOI: 10.3389/fimmu.2022.987771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/31/2022] [Indexed: 11/19/2022] Open
Abstract
The axons of retinal ganglion cells (RGCs) pass through the optic nerve head (ONH) and form the optic nerve (ON). The ONH serves as an anatomical interface between the vitreous cavity and subarachnoid space. After inducing acute neuroinflammation by intravitreal injection of lipopolysaccharides (LPS), we observed inflammatory activation in the retina, but detect no signs of inflammation in the posterior ON or infiltration of inflammatory cells in the ONH. Therefore, we hypothesized that the ONH functions as a barrier to vitreous inflammation. Using transmission electron microscopy, we identified significant increase in G-ratio in the posterior ON on day 7 post intravitreal injection (PII) of LPS compared with the phosphate buffered saline (PBS) group. Moreover, using confocal imaging of ex vivo tissue extracted from Aldh1L1-eGFP reporter mice, we observed that the ONH astrocytes altered their spatial orientation by elongating their morphology along the axonal axis of RGCs in LPS- versus PBS-treated eyes; this was quantified by the ratio of longitudinal (DL) and transverse (DT) diameter of astrocytes and the proportion of longitudinally locating astrocytes. Supportive evidences were further provided by transmission electron microscopic imaging in rat ONH. We further conducted RNA sequencing of ONH on day 1 PII and found LPS induced clear upregulation of immune and inflammatory pathways. Furthermore, gene set enrichment analysis revealed that astrocyte and microglia contributed prominently to the transcriptomic alterations in ONH. Here, we report that the vitreous infectious insults induce morphological changes of ONH astrocytes and transcriptomic alterations in the ONH. Glial responses in the ONH may defend against vitreous infectious insults and serve as a barrier to inflammation for the central nervous system.
Collapse
Affiliation(s)
- Wenwen Lai
- Department of Pharmacology, School of Basic Medical Sciences; Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Fudan University, Shanghai, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Huang
- Department of Pharmacology, School of Basic Medical Sciences; Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Fudan University, Shanghai, China
- Department of Ophthalmology, Eye, Ear, Nose and Throat Hospital; State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wangyi Fang
- Department of Ophthalmology, Shanghai Children’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Saiyue Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Qiao
- Department of Ophthalmology, Shanghai Children’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Gezhi Xu
- Department of Pharmacology, School of Basic Medical Sciences; Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Fudan University, Shanghai, China
- Department of Ophthalmology, Eye, Ear, Nose and Throat Hospital; State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xiaowei Wang
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, United States
- *Correspondence: Fengfei Ding, ; Xiaowei Wang,
| | - Fengfei Ding
- Department of Pharmacology, School of Basic Medical Sciences; Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Fudan University, Shanghai, China
- Department of Ophthalmology, Eye, Ear, Nose and Throat Hospital; State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- *Correspondence: Fengfei Ding, ; Xiaowei Wang,
| |
Collapse
|
20
|
Study on the Effect of Dexmedetomidine on Postoperative Cognitive Dysfunction and Inflammation in Aged Rats. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:8830706. [PMID: 35866065 PMCID: PMC9270136 DOI: 10.1155/2022/8830706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 12/26/2022]
Abstract
Objective The aim of this study is to investigate the effect of dexmedetomidine on cognitive dysfunction and inflammatory cytokines in the hippocampus after surgery in aged rats. Methods A total of 30 healthy male Sprague Dawley rats were divided into control group, sham group, and dexmedetomidine group. A splenectomy rat model was established and dexmedetomidine was intraperitoneally injected before operation. The cognitive function of rats was examined by Morris Water-Maze Test, open field experiment, and passive avoidance memory test. And the expression levels of IL-6, IL-1β, and TNF-α in the hippocampus were examined by ELISA. Results The escape latency for 5 continuous days in dexmedetomidine group was significantly decreased comparing with control group (all P < 0.05). The number of times of swimming and the percentage of swimming time in dexmedetomidine group were significantly more than those in control group (all P < 0.05). What is more, rats in dexmedetomidine group had the decreased time of stay in the central square and the increased number of standing times in comparison with the control group, and the statistical differences were found (all P < 0.05). Compared with the control group, dexmedetomidine intraperitoneally injected before surgery could significantly inhibit the expression levels of IL-6, IL-1β, and TNF-α in the hippocampus, and there were statistical differences (all P < 0.05). Conclusion Dexmedetomidine could significantly relieve the postoperative cognitive dysfunction in aged rats. The mechanism may be associated with the decreased inflammatory cytokines in the hippocampus.
Collapse
|
21
|
Wang T, Weng H, Zhou H, Yang Z, Tian Z, Xi B, Li Y. Esketamine alleviates postoperative depression-like behavior through anti-inflammatory actions in mouse prefrontal cortex. J Affect Disord 2022; 307:97-107. [PMID: 35378150 DOI: 10.1016/j.jad.2022.03.072] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 03/16/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023]
Abstract
The rising incidence of postoperative depression (POD) in recent years has placed a heavy burden on patients' physical and mental health. At this point in time, however, POD pathogenesis remains poorly understood and novel therapeutic strategies are being sought. The present study aimed to clarify esketamine's protective effects and possible mechanisms of action in POD. To this avail, we used an animal model of postoperative depression to analyze behavioral, parameters, plus the inflammatory response in serum and in the medial prefrontal cortex (mPFC). Using immunofluorescence staining, we detected the number of microglia and parvalbumin (PV) in mPFC, and determined changes in neuronal dendritic spine density via Golgi staining. Expression of Iba1, PSD95 and NF-κB was examined by Western blot analysis. Our results show that esketamine can significantly improve depression-like symptoms caused by anesthesia and surgery. In addition, esketamine administration reversed the decrease in the density of PV neurons and restored synaptogenesis in mPFC which had been perturbed by inflammation. The evidence obtained suggests esketamine's anti-inflammatory effects may be mediated by the BDNF/TrkB signaling pathway and possibly by attenuation of the nuclear factor κB (NF-κB) pathway. These data warrant further investigations into the interplay of esketamine, and microglia in the modulation of POD symptomatology.
Collapse
Affiliation(s)
- Tianyuan Wang
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Huandi Weng
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, Guangdong, China
| | - Hongji Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Zecheng Yang
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Zhongyou Tian
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China
| | - Biao Xi
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233003, Anhui, China
| | - Yalan Li
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China.
| |
Collapse
|
22
|
Wu YQ, Xiong J, He ZL, Yuan Y, Wang BN, Xu JY, Wu M, Zhang SS, Cai SF, Zhao JX, Xu K, Zhang HY, Xiao J. Metformin promotes microglial cells to facilitate myelin debris clearance and accelerate nerve repairment after spinal cord injury. Acta Pharmacol Sin 2022; 43:1360-1371. [PMID: 34480113 PMCID: PMC9160053 DOI: 10.1038/s41401-021-00759-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/08/2021] [Indexed: 02/07/2023]
Abstract
Spinal cord injury (SCI) is one kind of severe trauma for central nervous system. Myelin debris clearance and axon regeneration are essential for nerve regeneration after SCI. Metformin, a glucose-lowering drug, has been demonstrated to promote the locomotor functional recovery after SCI. In this study, we investigated the role and molecular mechanism of metformin on myelin preservation in a rat SCI model. SCI was induced in rats by compression at T9 level using a vascular clip. We showed that administration of metformin (50 mg·kg-1·d-1, ip) for 28 days significantly improved locomotor function in SCI rats. Metformin also ameliorated SCI-induced neuronal apoptosis and promoted axon regeneration in the spinal cord. Using co-immunofluorescence of IBa-1 and MBP, and luxol fasting blue (LFB) staining, we demonstrated that metformin promoted the transformation of M1 to M2 phenotype polarization of microglial cells, then greatly facilitated myelin debris clearance and protected the myelin in SCI rats. Furthermore, metformin ameliorated SCI-induced blockade of autophagic flux in the spinal cord, and enhanced the fusion of autophagosome and lysosome by inhibiting the AMPK-mTOR signaling pathway. Moreover, metformin significantly attenuated inflammatory responses in the spinal cord. In LPS-treated BV2 cells, pretreatment with metformin (2 mM) significantly enhanced autophagy level, suppressed inflammation and cell apoptosis. The protective effects were blocked in the presence of an autophagy inhibitor 3-methyladenine (3-MA, 5 mM), suggesting that the effect of metformin on autophagy in microglial cells is essential for the myelin preservation during nerve recovery. This study reveals a novel therapeutic effect of metformin in SCI recovery by regulating the activation of microglial cells and enhancing its autophagy level.
Collapse
Affiliation(s)
- Yan-Qing Wu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Jun Xiong
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Zi-Li He
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuan Yuan
- Department of Pharmacy, Hangzhou Red Cross Hospital, Zhejiang Province Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, 310003, China
| | - Bei-Ni Wang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Jing-Yu Xu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Man Wu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Su-Su Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Shu-Fang Cai
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Jia-Xin Zhao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Ke Xu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Hong-Yu Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China.
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
23
|
Mazzolini J, Le Clerc S, Morisse G, Coulonges C, Zagury J, Sieger D. Wasl is crucial to maintain microglial core activities during glioblastoma initiation stages. Glia 2022; 70:1027-1051. [PMID: 35194846 PMCID: PMC9306864 DOI: 10.1002/glia.24154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/28/2022]
Abstract
Microglia actively promotes the growth of high-grade gliomas. Within the glioma microenvironment an amoeboid microglial morphology has been observed, however the underlying causes and the related impact on microglia functions and their tumor promoting activities is unclear. Using the advantages of the larval zebrafish model, we identified the underlying mechanism and show that microglial morphology and functions are already impaired during glioma initiation stages. The presence of pre-neoplastic HRasV12 expressing cells induces an amoeboid morphology of microglia, increases microglial numbers and decreases their motility and phagocytic activity. RNA sequencing analysis revealed lower expression levels of the actin nucleation promoting factor wasla in microglia. Importantly, a microglia specific rescue of wasla expression restores microglial morphology and functions. This results in increased phagocytosis of pre-neoplastic cells and slows down tumor progression. In conclusion, we identified a mechanism that de-activates core microglial functions within the emerging glioma microenvironment. Restoration of this mechanism might provide a way to impair glioma growth.
Collapse
Affiliation(s)
- Julie Mazzolini
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Sigrid Le Clerc
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Gregoire Morisse
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Cédric Coulonges
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Jean‐François Zagury
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Dirk Sieger
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
24
|
Cook JR, Gray AL, Lemarchand E, Schiessl I, Green JP, Newland MC, Dyer DP, Brough D, Lawrence CB. LRRC8A is dispensable for a variety of microglial functions and response to acute stroke. Glia 2022; 70:1068-1083. [PMID: 35150591 PMCID: PMC9304177 DOI: 10.1002/glia.24156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/11/2022]
Abstract
Microglia, resident brain immune cells, are critical in orchestrating responses to central nervous system (CNS) injury. Many microglial functions, such as phagocytosis, motility and chemotaxis, are suggested to rely on chloride channels, including the volume‐regulated anion channel (VRAC), but studies to date have relied on the use of pharmacological tools with limited specificity. VRAC has also been proposed as a drug target for acute CNS injury, and its role in microglial function is of considerable interest for developing CNS therapeutics. This study aimed to definitively confirm the contribution of VRAC in microglia function by using conditional LRRC8A‐knockout mice, which lacked the essential VRAC subunit LRRC8A in microglia. We demonstrated that while VRAC contributed to cell volume regulation, it had no effect on phagocytic activity, cell migration or P2YR12‐dependent chemotaxis. Moreover, loss of microglial VRAC did not affect microglial morphology or the extent of ischemic damage following stroke. We conclude that VRAC does not critically regulate microglial responses to brain injury and could be targetable in other CNS cell types (e.g., astrocytes) without impeding microglial function. Our results also demonstrate a role for VRAC in cell volume regulation but show that VRAC is not involved in several major cellular functions that it was previously thought to regulate, and point to other, alternative mechanisms of chloride transport in innate immunity.
Collapse
Affiliation(s)
- James R Cook
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Anna L Gray
- Wellcome Centre for Cell-Matrix Research, Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Eloise Lemarchand
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ingo Schiessl
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jack P Green
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Mary C Newland
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Douglas P Dyer
- Wellcome Centre for Cell-Matrix Research, Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - David Brough
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catherine B Lawrence
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
25
|
Expression of the Adenosine A2A-A3 Receptor Heteromer in Different Brain Regions and Marked Upregulation in the Microglia of the Transgenic APPSw,Ind Alzheimer’s Disease Model. Biomedicines 2022; 10:biomedicines10020214. [PMID: 35203424 PMCID: PMC8869194 DOI: 10.3390/biomedicines10020214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Adenosine (Ado) receptors have been instrumental in the detection of heteromers and other higher-order receptor structures, mainly via interactions with other cell surface G-protein-coupled receptors. Apart from the first report of the A1 Ado receptor interacting with the A2A Ado receptor, there has been more recent data on the possibility that every Ado receptor type, A1, A2A, A2B, and A3, may interact with each other. The aim of this paper was to look for the expression and function of the A2A/A3 receptor heteromer (A2AA3Het) in neurons and microglia. In situ proximity ligation assays (PLA), performed in primary cells, showed that A2AA3Het expression was markedly higher in striatal than in cortical and hippocampal neurons, whereas it was similar in resting and activated microglia. Signaling assays demonstrated that the effect of the A2AR agonist, PSB 777, was reduced in the presence of the A3R agonist, 2-Cl-IB-MECA, whereas the effect of the A3R agonist was potentiated by the A2AR antagonist, SCH 58261. Interestingly, the expression of the heteromer was markedly enhanced in microglia from the APPSw,Ind model of Alzheimer’s disease. The functionality of the heteromer in primary microglia from APPSw,Ind mice was more similar to that found in resting microglia from control mice.
Collapse
|
26
|
Dyne E, Cawood M, Suzelis M, Russell R, Kim MH. Ultrastructural analysis of the morphological phenotypes of microglia associated with neuroinflammatory cues. J Comp Neurol 2021; 530:1263-1275. [PMID: 34773250 DOI: 10.1002/cne.25274] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/15/2022]
Abstract
Microglia are the primary resident immune cells of the central nervous system that are responsible for the maintenance of brain homeostasis. There is a plethora of evidence to suggest that microglia display distinct phenotypes that are associated with the alteration of cell morphology under varying environmental cues. However, it has not been fully explored how the varying states of microglial activation are linked to the alteration of microglia morphology, especially in the microdomain. The objective of this study was to quantitatively characterize the ultrastructural morphology of human microglia under neuroinflammatory cues. To address this, a human cell line of microglia was stimulated by antiinflammatory (IL-4), proinflammatory (TNF-α), and Alzheimer's disease (AD)-associated cues (Aβ, Aβ + TNF-α). The resulting effects on microglia morphology associated with changes in microdomain were analyzed using a high-resolution scanning electron microscopy. Our findings demonstrated that microglial activation under proinflammatory and AD-cues were closely linked to changes not only in cell shape but also in cell surface topography and higher-order branching of processes. Furthermore, our results revealed that microglia under proinflammatory cues exhibited unique morphological features involving cell-to-cell contact and the formation of vesicle-like structures. Our study provides insight into the fine details of microglia morphology associated with varying status of microglial activation.
Collapse
Affiliation(s)
- Eric Dyne
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
| | - Meghan Cawood
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Matthew Suzelis
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Reagan Russell
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Min-Ho Kim
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA.,Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| |
Collapse
|
27
|
Cordella F, Sanchini C, Rosito M, Ferrucci L, Pediconi N, Cortese B, Guerrieri F, Pascucci GR, Antonangeli F, Peruzzi G, Giubettini M, Basilico B, Pagani F, Grimaldi A, D’Alessandro G, Limatola C, Ragozzino D, Di Angelantonio S. Antibiotics Treatment Modulates Microglia-Synapses Interaction. Cells 2021; 10:cells10102648. [PMID: 34685628 PMCID: PMC8534187 DOI: 10.3390/cells10102648] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
‘Dysbiosis’ of the adult gut microbiota, in response to challenges such as infection, altered diet, stress, and antibiotics treatment has been recently linked to pathological alteration of brain function and behavior. Moreover, gut microbiota composition constantly controls microglia maturation, as revealed by morphological observations and gene expression analysis. However, it is unclear whether microglia functional properties and crosstalk with neurons, known to shape and modulate synaptic development and function, are influenced by the gut microbiota. Here, we investigated how antibiotic-mediated alteration of the gut microbiota influences microglial and neuronal functions in adult mice hippocampus. Hippocampal microglia from adult mice treated with oral antibiotics exhibited increased microglia density, altered basal patrolling activity, and impaired process rearrangement in response to damage. Patch clamp recordings at CA3-CA1 synapses revealed that antibiotics treatment alters neuronal functions, reducing spontaneous postsynaptic glutamatergic currents and decreasing synaptic connectivity, without reducing dendritic spines density. Antibiotics treatment was unable to modulate synaptic function in CX3CR1-deficient mice, pointing to an involvement of microglia–neuron crosstalk through the CX3CL1/CX3CR1 axis in the effect of dysbiosis on neuronal functions. Together, our findings show that antibiotic alteration of gut microbiota impairs synaptic efficacy, suggesting that CX3CL1/CX3CR1 signaling supporting microglia is a major player in in the gut–brain axis, and in particular in the gut microbiota-to-neuron communication pathway.
Collapse
Affiliation(s)
- Federica Cordella
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University, 00185 Rome, Italy; (F.C.); (C.S.); (L.F.); (B.B.); (G.D.); (D.R.)
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy; (N.P.); (G.P.); (F.P.); (A.G.)
| | - Caterina Sanchini
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University, 00185 Rome, Italy; (F.C.); (C.S.); (L.F.); (B.B.); (G.D.); (D.R.)
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy; (N.P.); (G.P.); (F.P.); (A.G.)
| | - Maria Rosito
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy; (N.P.); (G.P.); (F.P.); (A.G.)
- Correspondence: (M.R.); (S.D.A.)
| | - Laura Ferrucci
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University, 00185 Rome, Italy; (F.C.); (C.S.); (L.F.); (B.B.); (G.D.); (D.R.)
| | - Natalia Pediconi
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy; (N.P.); (G.P.); (F.P.); (A.G.)
| | - Barbara Cortese
- National Research Council-Nanotechnology Institute, 00185 Rome, Italy;
| | - Francesca Guerrieri
- Cancer Research Center of Lyon (CRCL), UMR Inserm U1052/CNRS 5286, 69373 Lyon, France; (F.G.); (G.R.P.)
| | - Giuseppe Rubens Pascucci
- Cancer Research Center of Lyon (CRCL), UMR Inserm U1052/CNRS 5286, 69373 Lyon, France; (F.G.); (G.R.P.)
| | - Fabrizio Antonangeli
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University, 00185 Rome, Italy;
| | - Giovanna Peruzzi
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy; (N.P.); (G.P.); (F.P.); (A.G.)
| | | | - Bernadette Basilico
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University, 00185 Rome, Italy; (F.C.); (C.S.); (L.F.); (B.B.); (G.D.); (D.R.)
| | - Francesca Pagani
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy; (N.P.); (G.P.); (F.P.); (A.G.)
- National Research Council-Nanotechnology Institute, 00185 Rome, Italy;
| | - Alfonso Grimaldi
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy; (N.P.); (G.P.); (F.P.); (A.G.)
| | - Giuseppina D’Alessandro
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University, 00185 Rome, Italy; (F.C.); (C.S.); (L.F.); (B.B.); (G.D.); (D.R.)
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University, 00185 Rome, Italy;
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University, 00185 Rome, Italy;
- IRCCS Neuromed, Via Atinese 18, 86077 Pozzilli, Italy
| | - Davide Ragozzino
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University, 00185 Rome, Italy; (F.C.); (C.S.); (L.F.); (B.B.); (G.D.); (D.R.)
- Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University, 00185 Rome, Italy; (F.C.); (C.S.); (L.F.); (B.B.); (G.D.); (D.R.)
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy; (N.P.); (G.P.); (F.P.); (A.G.)
- Correspondence: (M.R.); (S.D.A.)
| |
Collapse
|
28
|
Augusto-Oliveira M, Arrifano GP, Delage CI, Tremblay MÈ, Crespo-Lopez ME, Verkhratsky A. Plasticity of microglia. Biol Rev Camb Philos Soc 2021; 97:217-250. [PMID: 34549510 DOI: 10.1111/brv.12797] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023]
Abstract
Microglial cells are the scions of foetal macrophages which invade the neural tube early during embryogenesis. The nervous tissue environment instigates the phenotypic metamorphosis of foetal macrophages into idiosyncratic surveilling microglia, which are generally characterised by a small cell body and highly ramified motile processes that constantly scan the nervous tissue for signs of changes in homeostasis and allow microglia to perform crucial homeostatic functions. The surveilling microglial phenotype is evolutionarily conserved from early invertebrates to humans. Despite this evolutionary conservation, microglia show substantial heterogeneity in their gene and protein expression, as well as morphological appearance. These differences are age, region and context specific and reflect a high degree of plasticity underlying the life-long adaptation of microglia, supporting the exceptional adaptive capacity of the central nervous system. Microgliocytes are essential elements of cellular network formation and refinement in the developing nervous tissue. Several distinct patrolling modes of microglial processes contribute to the formation, modification, and pruning of synapses; to the support and protection of neurones through microglial-somatic junctions; and to the control of neuronal and axonal excitability by specific microglia-axonal contacts. In pathology, microglia undergo proliferation and reactive remodelling known as microgliosis, which is context dependent, yet represents an evolutionarily conserved defence response. Microgliosis results in the emergence of multiple disease and context-specific reactive states; in addition, neuropathology is associated with the appearance of specific protective or recovery microglial forms. In summary, the plasticity of microglia supports the development and functional activity of healthy nervous tissue and provides highly sophisticated defences against disease.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-110, Belém, Brazil
| | - Gabriela P Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-110, Belém, Brazil
| | - Charlotte Isabelle Delage
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, V8P 5C2, Canada.,Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec City, QC, G1V 4G2, Canada.,Neurology and Neurosurgery Department, McGill University, 3801 University Street, Montreal, QC, H3A 2B4, Canada.,Department of Molecular Medicine, Université Laval, Pavillon Ferdinand-Vandry, Bureau 4835, 1050 Avenue de la Médecine, Québec City, QC, G1V 0A6, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Life Sciences Center, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-110, Belém, Brazil
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, U.K.,Achucarro Center for Neuroscience, IKERBASQUE, 48011, Bilbao, Spain.,Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania
| |
Collapse
|
29
|
Wong EL, Strohm A, Atlas J, Lamantia C, Majewska AK. Dynamics of microglia and dendritic spines in early adolescent cortex after developmental alcohol exposure. Dev Neurobiol 2021; 81:786-804. [PMID: 34228891 PMCID: PMC8440400 DOI: 10.1002/dneu.22843] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/26/2021] [Accepted: 06/13/2021] [Indexed: 11/05/2022]
Abstract
Fetal alcohol spectrum disorder patients suffer from many cognitive disabilities. These include impaired auditory, visual, and tactile sensory information processing, making it more difficult for these patients to learn to navigate social scenarios. Rodent studies have shown that alcohol exposure during the brain growth spurt (BGS) can lead to acute neuronal apoptosis and an immunological response by microglia in the somatosensory cortex. Since microglia have critical physiological functions, including the support of excitatory synapse remodeling via interactions with dendritic spines, we sought to understand whether BGS alcohol exposure has long-term effects on microglial or dendritic spine dynamics. Using in vivo two-photon microscopy in 4-5 week old mice, we evaluated microglial functions such as process motility, the response to tissue injury, and the dynamics of physical interactions between microglial processes and dendritic spines. We also investigated potential differences in the morphology, density, or dynamics of dendritic spines in layer I/II primary sensory cortex of control and BGS alcohol exposed mice. We found that microglial process motility and contact with dendritic spines were not altered after BGS alcohol exposure. While the response of microglial processes toward tissue injury was not significantly altered by prior alcohol exposure, there was a trend suggesting that alcohol early in life may prime microglia to respond more quickly to secondary injury. Spine density, morphology, stability, and remodeling over time were not perturbed after BGS alcohol exposure. We demonstrate that after BGS alcohol exposure, the physiological functions of microglia and excitatory neurons remain intact in early adolescence.
Collapse
Affiliation(s)
- Elissa L. Wong
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Environmental Medicine, University of Rochester Medical Center, NY, USA
| | - Alexandra Strohm
- Department of Environmental Medicine, University of Rochester Medical Center, NY, USA
| | - Jason Atlas
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Cassandra Lamantia
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Ania K. Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
30
|
Odoj K, Brawek B, Asavapanumas N, Mojtahedi N, Heneka MT, Garaschuk O. In vivo mechanisms of cortical network dysfunction induced by systemic inflammation. Brain Behav Immun 2021; 96:113-126. [PMID: 34052361 DOI: 10.1016/j.bbi.2021.05.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/22/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022] Open
Abstract
Peripheral inflammation is known to impact brain function, resulting in lethargy, loss of appetite and impaired cognitive abilities. However, the channels for information transfer from the periphery to the brain, the corresponding signaling molecules and the inflammation-induced interaction between microglia and neurons remain obscure. Here, we used longitudinal in vivo two-photon Ca2+ imaging to monitor neuronal activity in the mouse cortex throughout the early (initiation) and late (resolution) phases of peripheral inflammation. Single peripheral lipopolysaccharide injection induced a substantial but transient increase in ongoing neuronal activity, restricted to the initiation phase, whereas the impairment of visual processing was selectively observed during the resolution phase of systemic inflammation. In the frontal/motor cortex, the initiation phase-specific cortical hyperactivity was seen in the deep (layer 5) and superficial (layer 2/3) pyramidal neurons but not in the axons coming from the somatosensory cortex, and was accompanied by reduced activity of layer 2/3 cortical interneurons. Moreover, the hyperactivity was preserved after depletion of microglia and in NLRP3-/- mice but absent in TNF-α-/- mice. Together, these data identify microglia-independent and TNF-α-mediated reduction of cortical inhibition as a likely cause of the initiation phase-specific cortical hyperactivity and reveal the resolution phase-specific impairment of sensory processing, presumably caused by activated microglia.
Collapse
Affiliation(s)
- Karin Odoj
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Bianca Brawek
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nithi Asavapanumas
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nima Mojtahedi
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Olga Garaschuk
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany.
| |
Collapse
|
31
|
Guo DH, Yamamoto M, Hernandez CM, Khodadadi H, Baban B, Stranahan AM. Beige adipocytes mediate the neuroprotective and anti-inflammatory effects of subcutaneous fat in obese mice. Nat Commun 2021; 12:4623. [PMID: 34330904 PMCID: PMC8324783 DOI: 10.1038/s41467-021-24540-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 06/24/2021] [Indexed: 12/25/2022] Open
Abstract
Visceral obesity increases risk of cognitive decline in humans, but subcutaneous adiposity does not. Here, we report that beige adipocytes are indispensable for the neuroprotective and anti-inflammatory effects of subcutaneous fat. Mice lacking functional beige fat exhibit accelerated cognitive dysfunction and microglial activation with dietary obesity. Subcutaneous fat transplantation also protects against chronic obesity in wildtype mice via beige fat-dependent mechanisms. Beige adipocytes restore hippocampal synaptic plasticity following transplantation, and these effects require the anti-inflammatory cytokine interleukin-4 (IL4). After observing beige fat-mediated induction of IL4 in meningeal T-cells, we investigated the contributions of peripheral lymphocytes in donor fat. There was no sign of donor-derived lymphocyte trafficking between fat and brain, but recipient-derived lymphocytes were required for the effects of transplantation on cognition and microglial morphology. These findings indicate that beige adipocytes oppose obesity-induced cognitive impairment, with a potential role for IL4 in the relationship between beige fat and brain function.
Collapse
Affiliation(s)
- De-Huang Guo
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Masaki Yamamoto
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Caterina M Hernandez
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Hesam Khodadadi
- Department of Oral Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Babak Baban
- Department of Oral Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Plastic Surgery Section, Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Alexis M Stranahan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
32
|
Voskanyan AV, Darbinyan AA, Parseghyan LM. Hemorrhagic changes and microglia activation induced by Macrovipera lebetina obtusa venom with the inhibited enzymatic activity in rat brain. Toxicol Res 2021; 38:195-204. [PMID: 35419270 PMCID: PMC8960507 DOI: 10.1007/s43188-021-00102-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/21/2021] [Accepted: 06/25/2021] [Indexed: 11/30/2022] Open
Abstract
The metalloproteinases and phospholipase A2 are the main enzymes in the venom of Macrovipera lebetina obtusa that play a decisive role in the destructive and toxic effects on the organism of the prey. Metalloproteinases cause hemorrhagic damage, destroy the basement membrane of the blood vessel and disrupt the connections between endothelial cells. Phospholipase A2 causes hemolysis of erythrocytes, destroy the cell membranes, and inhibits the adhesion of platelets and so on. The state of the capillaries of the rat brain and microglia under the action of the venom with separately inhibited enzymes was investigated and compared to the action of the crude venom. Also, the toxicity LD50 of the venom of Macrovipera lebetina obtusa with the inhibited enzymatic activity was determined. The histochemical study showed that the inhibition of phospholipase A2 enzymatic activity did not significantly change the vasodestructive effect of the venoms. In case of action of a venom with inhibited enzymatic activity of metalloproteinases, low activity of microglia and less damaged capillaries were observed. The toxicity of the venom with inhibited phospholipase A2 and with inhibited metalloproteinases was respectively 1.8 and 3.7 times weaker than that of the crude venom. We can claim that both the toxicity of the venom of Macrovipera lebetina obtusa, the damaged brain vessels and the increased activity of CNS microglia are determined mainly by the action of metalloproteinases.
Collapse
Affiliation(s)
- Armen V. Voskanyan
- Orbeli Institute of Physiology, National Academy of Sciences Armenia, 0028 Yerevan, Armenia
| | - Anna A. Darbinyan
- Orbeli Institute of Physiology, National Academy of Sciences Armenia, 0028 Yerevan, Armenia
| | - Lilya M. Parseghyan
- Orbeli Institute of Physiology, National Academy of Sciences Armenia, 0028 Yerevan, Armenia
| |
Collapse
|
33
|
Moraes CA, Zaverucha-do-Valle C, Fleurance R, Sharshar T, Bozza FA, d’Avila JC. Neuroinflammation in Sepsis: Molecular Pathways of Microglia Activation. Pharmaceuticals (Basel) 2021; 14:ph14050416. [PMID: 34062710 PMCID: PMC8147235 DOI: 10.3390/ph14050416] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/11/2022] Open
Abstract
Frequently underestimated, encephalopathy or delirium are common neurological manifestations associated with sepsis. Brain dysfunction occurs in up to 80% of cases and is directly associated with increased mortality and long-term neurocognitive consequences. Although the central nervous system (CNS) has been classically viewed as an immune-privileged system, neuroinflammation is emerging as a central mechanism of brain dysfunction in sepsis. Microglial cells are major players in this setting. Here, we aimed to discuss the current knowledge on how the brain is affected by peripheral immune activation in sepsis and the role of microglia in these processes. This review focused on the molecular pathways of microglial activity in sepsis, its regulatory mechanisms, and their interaction with other CNS cells, especially with neuronal cells and circuits.
Collapse
Affiliation(s)
- Carolina Araújo Moraes
- Immunopharmacology Lab, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21045-900, Brazil;
| | - Camila Zaverucha-do-Valle
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Ministry of Health, Rio de Janeiro 21040-360, Brazil; (C.Z.-d.-V.); (F.A.B.)
| | - Renaud Fleurance
- UCB Biopharma SRL, 1420 Braine L’Alleud, Belgium;
- Experimental Neuropathology, Infection, and Epidemiology Department, Institut Pasteur, 75015 Paris, France;
- Université de Paris Sciences et Lettres, 75006 Paris Paris, France
| | - Tarek Sharshar
- Experimental Neuropathology, Infection, and Epidemiology Department, Institut Pasteur, 75015 Paris, France;
- Neuro-Anesthesiology and Intensive Care Medicine, Sainte-Anne Hospital, Paris-Descartes University, 75015 Paris, France
| | - Fernando Augusto Bozza
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Ministry of Health, Rio de Janeiro 21040-360, Brazil; (C.Z.-d.-V.); (F.A.B.)
- D’Or Institute for Research and Education, Rio de Janeiro 22281-100, Brazil
| | - Joana Costa d’Avila
- Immunopharmacology Lab, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21045-900, Brazil;
- School of Medicine, Universidade Iguaçu, Rio de Janeiro 26260-045, Brazil
- Correspondence:
| |
Collapse
|
34
|
Shukla PK, Delotterie DF, Xiao J, Pierre JF, Rao R, McDonald MP, Khan MM. Alterations in the Gut-Microbial-Inflammasome-Brain Axis in a Mouse Model of Alzheimer's Disease. Cells 2021; 10:cells10040779. [PMID: 33916001 PMCID: PMC8067249 DOI: 10.3390/cells10040779] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD), a progressive neurodegenerative disorder characterized by memory loss and cognitive decline, is a major cause of death and disability among the older population. Despite decades of scientific research, the underlying etiological triggers are unknown. Recent studies suggested that gut microbiota can influence AD progression; however, potential mechanisms linking the gut microbiota with AD pathogenesis remain obscure. In the present study, we provided a potential mechanistic link between dysbiotic gut microbiota and neuroinflammation associated with AD progression. Using a mouse model of AD, we discovered that unfavorable gut microbiota are correlated with abnormally elevated expression of gut NLRP3 and lead to peripheral inflammasome activation, which in turn exacerbates AD-associated neuroinflammation. To this end, we observe significantly altered gut microbiota compositions in young and old 5xFAD mice compared to age-matched non-transgenic mice. Moreover, 5xFAD mice demonstrated compromised gut barrier function as evident from the loss of tight junction and adherens junction proteins compared to non-transgenic mice. Concurrently, we observed increased expression of NLRP3 inflammasome and IL-1β production in the 5xFAD gut. Consistent with our hypothesis, increased gut–microbial–inflammasome activation is positively correlated with enhanced astrogliosis and microglial activation, along with higher expression of NLRP3 inflammasome and IL-1β production in the brains of 5xFAD mice. These data indicate that the elevated expression of gut–microbial–inflammasome components may be an important trigger for subsequent downstream activation of inflammatory and potentially cytotoxic mediators, and gastrointestinal NLRP3 may promote NLRP3 inflammasome-mediated neuroinflammation. Thus, modulation of the gut microbiota may be a potential strategy for the treatment of AD-related neurological disorders in genetically susceptible hosts.
Collapse
Affiliation(s)
- Pradeep K. Shukla
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
- Correspondence: (P.K.S.); (M.M.K.); Tel.: 1-901-448-3180; Fax: 1-901-448-1662 (M.M.K.)
| | - David F. Delotterie
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.F.D.); (J.X.); (M.P.M.)
| | - Jianfeng Xiao
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.F.D.); (J.X.); (M.P.M.)
| | - Joseph F. Pierre
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - RadhaKrishna Rao
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Michael P. McDonald
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.F.D.); (J.X.); (M.P.M.)
- Department of Anatomy & Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Mohammad Moshahid Khan
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (D.F.D.); (J.X.); (M.P.M.)
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Center for Muscle, Metabolism, and Neuropathology, Division of Rehabilitation Sciences and Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Correspondence: (P.K.S.); (M.M.K.); Tel.: 1-901-448-3180; Fax: 1-901-448-1662 (M.M.K.)
| |
Collapse
|
35
|
Andoh M, Koyama R. Assessing Microglial Dynamics by Live Imaging. Front Immunol 2021; 12:617564. [PMID: 33763064 PMCID: PMC7982483 DOI: 10.3389/fimmu.2021.617564] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Microglia are highly dynamic in the brain in terms of their ability to migrate, proliferate, and phagocytose over the course of an individual's life. Real-time imaging is a useful tool to examine how microglial behavior is regulated and how it affects the surrounding environment. However, microglia are sensitive to environmental stimuli, so they possibly change their state during live imaging in vivo, mainly due to surgical damage, and in vitro due to various effects associated with culture conditions. Therefore, it is difficult to perform live imaging without compromising the properties of the microglia under physiological conditions. To overcome this barrier, various experimental conditions have been developed; recently, it has become possible to perform live imaging of so-called surveillant microglia in vivo, ex vivo, and in vitro, although there are various limitations. Now, we can choose in vivo, ex vivo, or in vitro live imaging systems according to the research objective. In this review, we discuss the advantages and disadvantages of each experimental system and outline the physiological significance and molecular mechanisms of microglial behavior that have been elucidated by live imaging.
Collapse
Affiliation(s)
- Megumi Andoh
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
36
|
Meng F, Sun N, Liu D, Jia J, Xiao J, Dai H. BCL2L13: physiological and pathological meanings. Cell Mol Life Sci 2021; 78:2419-2428. [PMID: 33201252 PMCID: PMC11073179 DOI: 10.1007/s00018-020-03702-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
BCL2L13 is a BCL2-like protein. It has been discovered for two decades, now on the way to be a hotspot of research with its physiological and pathological meanings found in recent years. Start with the pro-apoptotic activity, there have been reported consecutively that BCL2L13 could also induce mitochondrial fragmentation, inhibit cell death and promote mitophagy. Similar to BNIP3, BCL2L13 cannot be indiscriminately categorized into pro- or anti-apoptotic proteins. It anchors in the mitochondrial outer membrane, and expresses in various cells and tissues. This article reviews for the first time that BCL2L13 functions in physiological processes, such as growth and development and energy metabolism, and its dysregulation participating in pathological processes, including cancer, bacterial infection, cardiovascular diseases and degenerative diseases, suggesting its important roles in these events.
Collapse
Affiliation(s)
- Fei Meng
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China
| | - Naitong Sun
- Department of Hematology, the Third People's Hospital of Yancheng, Yancheng, 224001, China
| | - Dongyan Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China
| | - Jia Jia
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- University of Science and Technology of China, Hefei, 230026, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China
| | - Jun Xiao
- Department of Urology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Haiming Dai
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China.
| |
Collapse
|
37
|
Bsat S, Halaoui A, Kobeissy F, Moussalem C, El Houshiemy MN, Kawtharani S, Omeis I. Acute ischemic stroke biomarkers: a new era with diagnostic promise? Acute Med Surg 2021; 8:e696. [PMID: 34745637 PMCID: PMC8552525 DOI: 10.1002/ams2.696] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/15/2021] [Accepted: 09/01/2021] [Indexed: 01/14/2023] Open
Abstract
Stroke is considered as the first cause of neurological dysfunction and second cause of death worldwide. Recombinant tissue plasminogen activator is the only chemical treatment for ischemic stroke approved by the US Food and Drug Administration. It was the only standard of care for a long time with a very narrow therapeutic window, which usually ranges from 3 to 4.5 h of stroke onset; until 2015, when multiple trials demonstrated the benefit of mechanical thrombectomy during the first 6 h. In addition, recent trials showed that mechanical thrombectomy can be beneficial up to 24 h if the patients meet certain criteria including the presence of magnetic resonance imaging/computed tomography perfusion mismatch, which allows better selectivity and higher recruitment of eligible stroke patients. However, magnetic resonance imaging/computed tomography perfusion is not available in all stroke centers. Hence, physicians need other easy and available diagnostic tools to select stroke patients eligible for mechanical thrombectomy. Moreover, stroke management is still challenging for physicians, particularly those dealing with patients with "wake-up" stroke. The resulting brain tissue damage of ischemic stroke and the subsequent pathological processes are mediated by multiple molecular pathways that are modulated by inflammatory markers and post-transcriptional activity. A considerable number of published works suggest the role of inflammatory and cardiac brain-derived biomarkers (serum matrix metalloproteinase, thioredoxin, neuronal and glial markers, and troponin proteins) as well as different biomarkers including the emerging roles of microRNAs. In this review, we assess the accumulating evidence regarding the current status of acute ischemic stroke diagnostic biomarkers that could guide physicians for better management of stroke patients. Our review could give an insight into the roles of the different emerging markers and microRNAs that can be of high diagnostic value in patients with stroke. In fact, the field of stroke research, similar to the field of traumatic brain injury, is in immense need for novel biomarkers that can stratify diagnosis, prognosis, and therapy.
Collapse
Affiliation(s)
- Shadi Bsat
- Division of NeurosurgeryDepartment of SurgeryAmerican University of Beirut Medical CenterBeirutLebanon
| | - Adham Halaoui
- Division of NeurosurgeryDepartment of SurgeryAmerican University of Beirut Medical CenterBeirutLebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular GeneticsFaculty of MedicineAmerican University of BeirutBeirutLebanon
| | - Charbel Moussalem
- Division of NeurosurgeryDepartment of SurgeryAmerican University of Beirut Medical CenterBeirutLebanon
| | | | - Sarah Kawtharani
- Division of NeurosurgeryDepartment of SurgeryAmerican University of Beirut Medical CenterBeirutLebanon
| | - Ibrahim Omeis
- Division of NeurosurgeryDepartment of SurgeryAmerican University of Beirut Medical CenterBeirutLebanon
- Department of NeurosurgeryBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
38
|
Zeng W, Zhang C, Long Q, Li Y. Dexmedetomidine Alleviates LPS-Induced Neuronal Dysfunction by Modulating the AKT/GSK-3β/CRMP-2 Pathway in Hippocampal Neurons. Neuropsychiatr Dis Treat 2021; 17:671-680. [PMID: 33727816 PMCID: PMC7955869 DOI: 10.2147/ndt.s297365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Dexmedetomidine, an α2-adrenergic receptor agonist, mitigates cognitive dysfunction in elderly patients after surgery with general anesthesia. However, the underlying mechanism by which dexmedetomidine reduces cognitive dysfunction remains to be fully elucidated. The aim of this study was to investigate the effects of dexmedetomidine on lipopolysaccharide (LPS)-induced neuronal dysfunction in cultured hippocampal neurons. METHODS LPS, in the presence and absence of dexmedetomidine, was applied to cultured hippocampal neurons to mimic post-surgical inflammation. Neuronal morphology, including neurite outgrowth and synaptic transmission, was observed, and miniature excitatory postsynaptic currents were recorded by electrophysiological patch-clamp. RESULTS LPS significantly impaired neurite outgrowth in hippocampal neurons in a concentration- and time-dependent manner, which was reversed by dexmedetomidine treatment. Electrophysiological patch-clamp results showed that LPS induced synaptic transmission dysfunction, which was restored after dexmedetomidine addition. Furthermore, Western blotting assays showed that LPS suppressed the AKT/GSK-3β/CRMP-2 signaling pathway and dexmedetomidine countered the inhibitory effect of LPS by re-activating this pathway. CONCLUSION In general, dexmedetomidine protected against the effects of LPS-induced hippocampal neuron damage, including neurite outgrowth and synaptic transmission. Overall, dexmedetomidine modulated the AKT/GSK-3β/CRMP-2 signaling pathway to alleviate LPS-induced neurological dysfunction.
Collapse
Affiliation(s)
- Wei Zeng
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China.,Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Zhongshan, 528400, Guangdong, People's Republic of China
| | - Chunyuan Zhang
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Zhongshan, 528400, Guangdong, People's Republic of China
| | - Qingshan Long
- Department of Neurosurgery, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, 516002, Guangdong, People's Republic of China
| | - Yalan Li
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
| |
Collapse
|
39
|
Merlini M, Rafalski VA, Ma K, Kim KY, Bushong EA, Rios Coronado PE, Yan Z, Mendiola AS, Sozmen EG, Ryu JK, Haberl MG, Madany M, Sampson DN, Petersen MA, Bardehle S, Tognatta R, Dean T, Acevedo RM, Cabriga B, Thomas R, Coughlin SR, Ellisman MH, Palop JJ, Akassoglou K. Microglial G i-dependent dynamics regulate brain network hyperexcitability. Nat Neurosci 2020; 24:19-23. [PMID: 33318667 PMCID: PMC8118167 DOI: 10.1038/s41593-020-00756-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/11/2020] [Indexed: 12/19/2022]
Abstract
Microglial surveillance is a key feature of brain physiology and disease. We found that Gi-dependent microglial dynamics prevent neuronal network hyperexcitability. By generating MgPTX mice to genetically inhibit Gi in microglia, we showed that sustained reduction of microglia brain surveillance and directed process motility induced spontaneous seizures and increased hypersynchrony upon physiologically evoked neuronal activity in awake adult mice. Thus, Gi-dependent microglia dynamics may prevent hyperexcitability in neurological diseases.
Collapse
Affiliation(s)
| | | | - Keran Ma
- Gladstone Institutes, San Francisco, CA, USA
| | - Keun-Young Kim
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.,National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Eric A Bushong
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.,National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | | | - Zhaoqi Yan
- Gladstone Institutes, San Francisco, CA, USA
| | | | - Elif G Sozmen
- Gladstone Institutes, San Francisco, CA, USA.,Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jae Kyu Ryu
- Gladstone Institutes, San Francisco, CA, USA.,Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Matthias G Haberl
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.,National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Matthew Madany
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.,National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Daniel Naranjo Sampson
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.,National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Mark A Petersen
- Gladstone Institutes, San Francisco, CA, USA.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Terry Dean
- Gladstone Institutes, San Francisco, CA, USA.,Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | | - Shaun R Coughlin
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Mark H Ellisman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.,National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Jorge J Palop
- Gladstone Institutes, San Francisco, CA, USA.,Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institutes, San Francisco, CA, USA. .,Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
40
|
Maternal immune activation induces sustained changes in fetal microglia motility. Sci Rep 2020; 10:21378. [PMID: 33288794 PMCID: PMC7721716 DOI: 10.1038/s41598-020-78294-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Maternal infection or inflammation causes abnormalities in brain development associated with subsequent cognitive impairment and in an increased susceptibility to schizophrenia and autism spectrum disorders. Maternal immune activation (MIA) and increases in serum cytokine levels mediates this association via effects on the fetal brain, and microglia can respond to maternal immune status, but consensus on how microglia may respond is lacking and no-one has yet examined if microglial process motility is impaired. In this study we investigated how MIA induced at two different gestational ages affected microglial properties at different developmental stages. Immune activation in mid-pregnancy increased IL-6 expression in embryonic microglia, but failed to cause any marked changes in morphology either at E18 or postnatally. In contrast MIA, particularly when induced earlier (at E12), caused sustained alterations in the patterns of microglial process motility and behavioral deficits. Our research has identified an important microglial property that is altered by MIA and which may contribute to the underlying pathophysiological mechanisms linking maternal immune status to subsequent risks for cognitive disease.
Collapse
|
41
|
Cserép C, Pósfai B, Dénes Á. Shaping Neuronal Fate: Functional Heterogeneity of Direct Microglia-Neuron Interactions. Neuron 2020; 109:222-240. [PMID: 33271068 DOI: 10.1016/j.neuron.2020.11.007] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/13/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
The functional contribution of microglia to normal brain development, healthy brain function, and neurological disorders is increasingly recognized. However, until recently, the nature of intercellular interactions mediating these effects remained largely unclear. Recent findings show microglia establishing direct contact with different compartments of neurons. Although communication between microglia and neurons involves intermediate cells and soluble factors, direct membrane contacts enable a more precisely regulated, dynamic, and highly effective form of interaction for fine-tuning neuronal responses and fate. Here, we summarize the known ultrastructural, molecular, and functional features of direct microglia-neuron interactions and their roles in brain disease.
Collapse
Affiliation(s)
- Csaba Cserép
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary
| | - Balázs Pósfai
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary; Szentágothai János Doctoral School of Neurosciences, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary.
| |
Collapse
|
42
|
Onodera J, Nagata H, Nakashima A, Ikegaya Y, Koyama R. Neuronal brain-derived neurotrophic factor manipulates microglial dynamics. Glia 2020; 69:890-904. [PMID: 33119934 DOI: 10.1002/glia.23934] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 10/22/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
Abstract
Brain-derived neurotrophic factor (BDNF), a main member of the neurotrophin family that is active in the brain, supports neuronal survival and growth. Microglial BDNF affects both the structural and functional properties of neurons. In contrast, whether and how neuronal BDNF affects microglial dynamics remain largely undetermined. Here, we examined the effects of BDNF on the properties of microglia in the CA3 region of the hippocampus. We chose this site because the axonal boutons of hippocampal mossy fibers, which are mostly formed in the CA3 region, contain the highest levels of BDNF in the rodent brain. We transfected mouse dentate granule cells with an adeno-associated virus that encodes both a BDNF short hairpin RNA (shRNA) and red fluorescent protein to examine the effects of mossy fiber-derived BDNF on microglia. Based on immunohistochemistry, BDNF knockdown with an shRNA resulted in an increase in microglial density in the mossy fiber pathway and increased engulfment of mossy fiber axons by microglia. In addition, we performed time-lapse imaging of microglial processes in hippocampal slice cultures to examine the effects of BDNF on microglial motility. Time-lapse imaging revealed increases in the motility of microglial processes and the engulfment of mossy fiber synapses by microglia when BDNF signaling was pharmacologically blocked. Thus, neuronal BDNF prevents microglia from engulfing mossy fiber synapses in the hippocampus.
Collapse
Affiliation(s)
- Junya Onodera
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidetaka Nagata
- Platform Technology Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Ai Nakashima
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
43
|
Won E, Kim YK. Neuroinflammation-Associated Alterations of the Brain as Potential Neural Biomarkers in Anxiety Disorders. Int J Mol Sci 2020; 21:ijms21186546. [PMID: 32906843 PMCID: PMC7555994 DOI: 10.3390/ijms21186546] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/30/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
Stress-induced changes in the immune system, which lead to neuroinflammation and consequent brain alterations, have been suggested as possible neurobiological substrates of anxiety disorders, with previous literature predominantly focusing on panic disorder, agoraphobia, and generalized anxiety disorder, among the anxiety disorders. Anxiety disorders have frequently been associated with chronic stress, with chronically stressful situations being reported to precipitate the onset of anxiety disorders. Also, chronic stress has been reported to lead to hypothalamic–pituitary–adrenal axis and autonomic nervous system disruption, which may in turn induce systemic proinflammatory conditions. Preliminary evidence suggests anxiety disorders are also associated with increased inflammation. Systemic inflammation can access the brain, and enhance pro-inflammatory cytokine levels that have been shown to precipitate direct and indirect neurotoxic effects. Prefrontal and limbic structures are widely reported to be influenced by neuroinflammatory conditions. In concordance with these findings, various imaging studies on panic disorder, agoraphobia, and generalized anxiety disorder have reported alterations in structure, function, and connectivity of prefrontal and limbic structures. Further research is needed on the use of inflammatory markers and brain imaging in the early diagnosis of anxiety disorders, along with the possible efficacy of anti-inflammatory interventions on the prevention and treatment of anxiety disorders.
Collapse
Affiliation(s)
- Eunsoo Won
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea;
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, Korea
- Correspondence: ; Tel.: +82-31-412-5140; Fax: +82-31-412-5144
| |
Collapse
|
44
|
Mitochondrial Transplantation Attenuates Brain Dysfunction in Sepsis by Driving Microglial M2 Polarization. Mol Neurobiol 2020; 57:3875-3890. [PMID: 32613465 DOI: 10.1007/s12035-020-01994-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
Activation of microglia and mitochondrial dysfunction are two major contributors to the pathogenesis of sepsis-associated brain dysfunction. Mitochondrial dysfunction can alter the immunological profile of microglia favoring to a pro-inflammatory phenotype. Mitochondrial transplantation, as an emerging mitochondria-targeted therapy, possesses considerable therapeutic potential in various central nervous system injuries or diseases. However, the effects of mitochondrial transplantation on microglial polarization and neuroprotection after sepsis remain unclear. In this study, lipopolysaccharide (LPS)/interferon-γ (IFN-γ) and interleukin-4 (IL-4)/interleukin-13 (IL-13) were used to induce different phenotypes of BV2 microglial cells. We observed that mitochondrial content and function were enhanced in IL-4-/IL-13-stimulated microglia. In vitro, mitochondria treatment conferred neuroprotection by enhancing microglial polarization from the M1 phenotype to the M2 phenotype and suppressing microglial-derived inflammatory cytokine release. Furthermore, microglial phenotypes and behavior tests were assessed after mice were subjected to sepsis by cecal ligation and puncture (CLP) followed by intracerebroventricular injection of exogenous functional mitochondria. We found that mitochondrial transplantation induced microglial M2 rather than M1 response 24 h after sepsis. Mitochondrial transplantation improved behavioral deficits by increasing the latency time in inhibitory avoidance test and decreasing the number of crossing and rearing in the test session of open field test 10 days after CLP onset. These findings indicate that mitochondrial transplantation promotes the phenotypic conversion of microglia and improves cognitive impairment in sepsis survivors, supporting the potential use of exogenous mitochondrial transplantation therapy that may be a potential therapeutic opportunity for sepsis-associated brain dysfunction.
Collapse
|
45
|
In vivo characterization of functional states of cortical microglia during peripheral inflammation. Brain Behav Immun 2020; 87:243-255. [PMID: 31837418 DOI: 10.1016/j.bbi.2019.12.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022] Open
Abstract
Peripheral inflammation is known to trigger a mirror inflammatory response in the brain, involving brain's innate immune cells - microglia. However, the functional phenotypes, which these cells adopt in the course of peripheral inflammation, remain obscure. In vivo two-photon imaging of microglial Ca2+ signaling as well as process motility reveals two distinct functional states of cortical microglia during a lipopolysaccharide-induced peripheral inflammation: an early "sensor state" characterized by dramatically increased intracellular Ca2+ signaling but ramified morphology and a later "effector state" characterized by slow normalization of intracellular Ca2+ signaling but hypertrophic morphology, substantial IL-1β production in a subset of cells as well as increased velocity of directed process extension and loss of coordination between individual processes. Thus, lipopolysaccharide-induced microglial Ca2+ signaling might represent the central element connecting receptive and executive functions of microglia.
Collapse
|
46
|
Uemura MT, Maki T, Ihara M, Lee VMY, Trojanowski JQ. Brain Microvascular Pericytes in Vascular Cognitive Impairment and Dementia. Front Aging Neurosci 2020; 12:80. [PMID: 32317958 PMCID: PMC7171590 DOI: 10.3389/fnagi.2020.00080] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Pericytes are unique, multi-functional mural cells localized at the abluminal side of the perivascular space in microvessels. Originally discovered in 19th century, pericytes had drawn less attention until decades ago mainly due to lack of specific markers. Recently, however, a growing body of evidence has revealed that pericytes play various important roles: development and maintenance of blood–brain barrier (BBB), regulation of the neurovascular system (e.g., vascular stability, vessel formation, cerebral blood flow, etc.), trafficking of inflammatory cells, clearance of toxic waste products from the brain, and acquisition of stem cell-like properties. In the neurovascular unit, pericytes perform these functions through coordinated crosstalk with neighboring cells including endothelial, glial, and neuronal cells. Dysfunction of pericytes contribute to a wide variety of diseases that lead to cognitive impairments such as cerebral small vessel disease (SVD), acute stroke, Alzheimer’s disease (AD), and other neurological disorders. For instance, in SVDs, pericyte degeneration leads to microvessel instability and demyelination while in stroke, pericyte constriction after ischemia causes a no-reflow phenomenon in brain capillaries. In AD, which shares some common risk factors with vascular dementia, reduction in pericyte coverage and subsequent microvascular impairments are observed in association with white matter attenuation and contribute to impaired cognition. Pericyte loss causes BBB-breakdown, which stagnates amyloid β clearance and the leakage of neurotoxic molecules into the brain parenchyma. In this review, we first summarize the characteristics of brain microvessel pericytes, and their roles in the central nervous system. Then, we focus on how dysfunctional pericytes contribute to the pathogenesis of vascular cognitive impairment including cerebral ‘small vessel’ and ‘large vessel’ diseases, as well as AD. Finally, we discuss therapeutic implications for these disorders by targeting pericytes.
Collapse
Affiliation(s)
- Maiko T Uemura
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,JSPS Overseas Research Fellowship Program, Japan Society for the Promotion of Science, Tokyo, Japan
| | - Takakuni Maki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Virginia M Y Lee
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - John Q Trojanowski
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
47
|
Identification of a pro-elongation effect of diallyl disulfide, a major organosulfur compound in garlic oil, on microglial process. J Nutr Biochem 2020; 78:108323. [DOI: 10.1016/j.jnutbio.2019.108323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 10/30/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022]
|
48
|
Kyrargyri V, Madry C, Rifat A, Arancibia‐Carcamo IL, Jones SP, Chan VTT, Xu Y, Robaye B, Attwell D. P2Y 13 receptors regulate microglial morphology, surveillance, and resting levels of interleukin 1β release. Glia 2020; 68:328-344. [PMID: 31520551 PMCID: PMC6916289 DOI: 10.1002/glia.23719] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 12/19/2022]
Abstract
Microglia sense their environment using an array of membrane receptors. While P2Y12 receptors are known to play a key role in targeting directed motility of microglial processes to sites of damage where ATP/ADP is released, little is known about the role of P2Y13 , which transcriptome data suggest is the second most expressed neurotransmitter receptor in microglia. We show that, in patch-clamp recordings in acute brain slices from mice lacking P2Y13 receptors, the THIK-1 K+ current density evoked by ADP activating P2Y12 receptors was increased by ~50%. This increase suggested that the P2Y12 -dependent chemotaxis response should be potentiated; however, the time needed for P2Y12 -mediated convergence of microglial processes onto an ADP-filled pipette or to a laser ablation was longer in the P2Y13 KO. Anatomical analysis showed that the density of microglia was unchanged, but that they were less ramified with a shorter process length in the P2Y13 KO. Thus, chemotactic processes had to grow further and so arrived later at the target, and brain surveillance was reduced by ~30% in the knock-out. Blocking P2Y12 receptors in brain slices from P2Y13 KO mice did not affect surveillance, demonstrating that tonic activation of these high-affinity receptors is not needed for surveillance. Strikingly, baseline interleukin-1β release was increased fivefold while release evoked by LPS and ATP was not affected in the P2Y13 KO, and microglia in intact P2Y13 KO brains were not detectably activated. Thus, P2Y13 receptors play a role different from that of their close relative P2Y12 in regulating microglial morphology and function.
Collapse
Affiliation(s)
- Vasiliki Kyrargyri
- Department of Neuroscience, Physiology, & PharmacologyUniversity College LondonLondonUK
- Department of Immunology, Laboratory of Molecular GeneticsHellenic Pasteur InstituteAthensGreece
| | - Christian Madry
- Department of Neuroscience, Physiology, & PharmacologyUniversity College LondonLondonUK
- Institute of NeurophysiologyCharité – Universitätsmedizin BerlinBerlinGermany
| | - Ali Rifat
- Institute of NeurophysiologyCharité – Universitätsmedizin BerlinBerlinGermany
| | | | - Steffan P. Jones
- Department of Neuroscience, Physiology, & PharmacologyUniversity College LondonLondonUK
| | - Victor T. T. Chan
- Department of Neuroscience, Physiology, & PharmacologyUniversity College LondonLondonUK
| | - Yajing Xu
- Department of Neuroscience, Physiology, & PharmacologyUniversity College LondonLondonUK
| | - Bernard Robaye
- Faculté de MédecineUniversité Libre de BruxellesBruxellesBelgium
| | - David Attwell
- Department of Neuroscience, Physiology, & PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
49
|
Meng F, Guo Z, Hu Y, Mai W, Zhang Z, Zhang B, Ge Q, Lou H, Guo F, Chen J, Duan S, Gao Z. CD73-derived adenosine controls inflammation and neurodegeneration by modulating dopamine signalling. Brain 2020; 142:700-718. [PMID: 30689733 DOI: 10.1093/brain/awy351] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/09/2018] [Accepted: 11/22/2018] [Indexed: 12/21/2022] Open
Abstract
Ectonucleotidase-mediated ATP catabolism provides a powerful mechanism to control the levels of extracellular adenosine. While increased adenosine A2A receptor (A2AR) signaling has been well-documented in both Parkinson's disease models and patients, the source of this enhanced adenosine signalling remains unclear. Here, we show that the ecto-5'-nucleotidase (CD73)-mediated adenosine formation provides an important input to activate A2AR, and upregulated CD73 and A2AR in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease models coordinatively contribute to the elevated adenosine signalling. Importantly, we demonstrate that CD73-derived adenosine-A2AR signalling modulates microglial immunoresponses and morphological dynamics. CD73 inactivation significantly attenuated lipopolysaccharide-induced pro-inflammatory responses in microglia, but enhanced microglia process extension, movement and morphological transformation in the laser injury and acute MPTP-induced Parkinson's disease models. Limiting CD73-derived adenosine substantially suppressed microglia-mediated neuroinflammation and improved the viability of dopaminergic neurons and motor behaviours in Parkinson's disease models. Moreover, CD73 inactivation suppressed A2AR induction and A2AR-mediated pro-inflammatory responses, whereas replenishment of adenosine analogues restored these effects, suggesting that CD73 produces a self-regulating feed-forward adenosine formation to activate A2AR and promote neuroinflammation. We further provide the first evidence that A2A enhanced inflammation by antagonizing dopamine-mediated anti-inflammation, suggesting that the homeostatic balance between adenosine and dopamine signalling is key to microglia immunoresponses. Our study thus reveals a novel role for CD73-mediated nucleotide metabolism in regulating neuroinflammation and provides the proof-of-principle that targeting nucleotide metabolic pathways to limit adenosine production and neuroinflammation in Parkinson's disease might be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Fan Meng
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhige Guo
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaling Hu
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Weihao Mai
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenjie Zhang
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Zhang
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianqian Ge
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Huifang Lou
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Guo
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiangfan Chen
- Molecular Neuropharmacology Laboratory and State Key Laboratory of Optometry, Ophthalmology and Vision Science, School of Optometry and Ophthalmology, Wenzhou, Zhejiang, China
| | - Shumin Duan
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Gao
- Department of Neurobiology and Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
50
|
Distinct P2Y Receptors Mediate Extension and Retraction of Microglial Processes in Epileptic and Peritumoral Human Tissue. J Neurosci 2020; 40:1373-1388. [PMID: 31896671 DOI: 10.1523/jneurosci.0218-19.2019] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Microglia exhibit multiple, phenotype-dependent motility patterns often triggered by purinergic stimuli. However, little data exist on motility of human microglia in pathological situations. Here we examine motility of microglia stained with a fluorescent lectin in tissue slices from female and male epileptic patients diagnosed with mesial temporal lobe epilepsy or cortical glioma (peritumoral cortex). Microglial shape varied from ramified to amoeboid cells predominantly in regions of high neuronal loss or closer to a tumor. Live imaging revealed unstimulated or purine-induced microglial motilities, including surveillance movements, membrane ruffling, and process extension or retraction. At different concentrations, ADP triggered opposing motilities. Low doses triggered process extension. It was suppressed by P2Y12 receptor antagonists, which also reduced process length and surveillance movements. Higher purine doses caused process retraction and membrane ruffling, which were blocked by joint application of P2Y1 and P2Y13 receptor antagonists. Purinergic effects on motility were similar for all microglia tested. Both amoeboid and ramified cells from mesial temporal lobe epilepsy or peritumoral cortex tissue expressed P2Y12 receptors. A minority of microglia expressed the adenosine A2A receptor, which has been linked with process withdrawal of rodent cells. Laser-mediated tissue damage let us test the functional significance of these effects. Moderate damage induced microglial process extension, which was blocked by P2Y12 receptor antagonists. Overall, the purine-induced motility of human microglia in epileptic tissue is similar to that of rodent microglia in that the P2Y12 receptor initiates process extension. It differs in that retraction is triggered by joint activation of P2Y1/P2Y13 receptors.SIGNIFICANCE STATEMENT Microglial cells are brain-resident immune cells with multiple functions in healthy or diseased brains. These diverse functions are associated with distinct phenotypes, including different microglial shapes. In the rodent, purinergic signaling is associated with changes in cell shape, such as process extension toward tissue damage. However, there are little data on living human microglia, especially in diseased states. We developed a reliable technique to stain microglia from epileptic and glioma patients to examine responses to purines. Low-intensity purinergic stimuli induced process extension, as in rodents. In contrast, high-intensity stimuli triggered a process withdrawal mediated by both P2Y1 and P2Y13 receptors. P2Y1/P2Y13 receptor activation has not previously been linked to microglial morphological changes.
Collapse
|