1
|
Alami K, Fathollahi Y, Hashemizadeh S, Mosleh M, Semnanian S, Mousavi SY, Azizi H. Microglia-dependent peripheral neuropathic pain in adulthood following adolescent exposure to morphine in male rats. Neuropharmacology 2025; 263:110211. [PMID: 39521039 DOI: 10.1016/j.neuropharm.2024.110211] [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: 08/07/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Persistent effects of adolescent morphine exposure on neurobiological processes and behaviors in adulthood have been partially identified. Hypersensitivity following adolescent exposure to morphine is a complex and multifaceted phenomenon whose underlying mechanisms remain largely unknown. This study aimed to investigate the involvement of microglia in neuropathic pain sensitivity following adolescent morphine exposure, focused on hippocampal genes expression and plasticity. To achieve this, adolescent male Wistar rats received morphine, along with minocycline, to inhibit microglial activity. The allodynia and hyperalgesia of adult rats were evaluated using von-Frey filaments and the Hargreaves plantar test in both baseline and neuropathic pain conditions. Hippocampal genes expression was analyzed following the behavioral tests. The plasticity of the Schaffer-CA1 hippocampal synapses was also assessed using field potential recording following neuropathy. Results showed that adolescent morphine exposure exacerbated the allodynia and hyperalgesia in both baseline and neuropathic pain states in adult rats, which was significantly reduced by the co-administration of minocycline during adolescence. Neuropathy in adult rats was found to increase hippocampal expression of inflammatory mediators, but adolescent morphine prevented this effect. Additionally, we observed a reduction in the baseline synaptic transmission and long-term potentiation (LTP) at the Schaffer-CA1 hippocampal synapses after neuropathy in adult rats following adolescent exposure to morphine. The reduction of synaptic activity was not altered by the co-administration of minocycline with morphine during adolescence. It is concluded that microglia play an important role in mediating hypersensitivity induced by adolescent morphine exposure, although hippocampal microglia may not be directly involved in this process.
Collapse
Affiliation(s)
- Kawsar Alami
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yaghoub Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shiva Hashemizadeh
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Masoumeh Mosleh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran
| | - Sayed Yousof Mousavi
- Department of Cognitive Neuroscience, Neuroscience Research Center, Kavosh Nonprofit Educational Research Institute, Kabul, Afghanistan
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
2
|
Konat GW. Neuroplasticity elicited by peripheral immune challenge with a viral mimetic. Brain Res 2025; 1846:149239. [PMID: 39284559 DOI: 10.1016/j.brainres.2024.149239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Peripheral viral infections are well known to profoundly alter brain function; however detailed mechanisms of this immune-to-brain communication have not been deciphered. This review focuses on studies of cerebral effects of peripheral viral challenge employing intraperitoneal injection of a viral mimetic, polyinosinic-polycytidylic acid (PIC). In this paradigm, PIC challenge induces the acute phase response (APR) characterized by a transient surge of circulating inflammatory factors, primarily IFNβ, IL-6 and CXCL10. The blood-borne factors, in turn, elicit the generation of CXCL10 by hippocampal neurons. Neurons also express the cognate receptor of CXCL10, i.e., CXCR3 implicating the existence of autocrine/paracrine signaling. The CXCL10/CXCR3 axis mediates the ensuing neuroplastic changes manifested as neuronal hyperexcitability, seizure hypersusceptibility, and sickness behavior. Electrophysiological studies revealed that the neuroplastic changes entail the potentiation of excitatory synapses likely at both pre- and postsynaptic loci. Excitatory synaptic transmission is further augmented by PIC challenge-induced elevation of extracellular glutamate that is mediated by astrocytes. In addition, the hyperexcitability of neuronal circuits might involve the repression of inhibitory signaling. Accordingly, CXCL10 released by neurons activates microglia whose processes invade perisomatic inhibitory synapses, resulting in a partial detachment of the presynaptic terminals, and thus, de-inhibition. This process might be facilitated by the cerebral complement system, which is also upregulated and activated by PIC challenge. Moreover, CXCL10 stimulates the expression of neuronal c-fos protein, another index of hyperexcitability. The reviewed studies form a foundation for full elucidation of the fascinating intersection between peripheral viral infections and neuroplasticity. Because the activation of such pathways may constitute a serious comorbidity factor for neuropathological conditions, this research would advance the development of preventive strategies.
Collapse
Affiliation(s)
- Gregory W Konat
- Department of Biochemistry and Molecular Medicine, Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV 26506, USA.
| |
Collapse
|
3
|
León-Rodríguez A, Grondona JM, Marín-Wong S, López-Aranda MF, López-Ávalos MD. Long-term reprogramming of primed microglia after moderate inhibition of CSF1R signaling. Glia 2025; 73:175-195. [PMID: 39448548 DOI: 10.1002/glia.24627] [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: 05/24/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024]
Abstract
In acute neuroinflammation, microglia activate transiently, and return to a resting state later on. However, they may retain immune memory of such event, namely priming. Primed microglia are more sensitive to new stimuli and develop exacerbated responses, representing a risk factor for neurological disorders with an inflammatory component. Strategies to control the hyperactivation of microglia are, hence, of great interest. The receptor for colony stimulating factor 1 (CSF1R), expressed in myeloid cells, is essential for microglia viability, so its blockade with specific inhibitors (e.g. PLX5622) results in significant depletion of microglial population. Interestingly, upon inhibitor withdrawal, new naïve microglia repopulate the brain. Depletion-repopulation has been proposed as a strategy to reprogram microglia. However, substantial elimination of microglia is inadvisable in human therapy. To overcome such drawback, we aimed to reprogram long-term primed microglia by CSF1R partial inhibition. Microglial priming was induced in mice by acute neuroinflammation, provoked by intracerebroventricular injection of neuraminidase. After 3-weeks recovery, low-dose PLX5622 treatment was administrated for 12 days, followed by a withdrawal period of 7 weeks. Twelve hours before euthanasia, mice received a peripheral lipopolysaccharide (LPS) immune challenge, and the subsequent microglial inflammatory response was evaluated. PLX5622 provoked a 40%-50% decrease in microglial population, but basal levels were restored 7 weeks later. In the brain regions studied, hippocampus and hypothalamus, LPS induced enhanced microgliosis and inflammatory activation in neuraminidase-injected mice, while PLX5622 treatment prevented these changes. Our results suggest that PLX5622 used at low doses reverts microglial priming and, remarkably, prevents broad microglial depletion.
Collapse
Affiliation(s)
- Ana León-Rodríguez
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - Jesús M Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - Sonia Marín-Wong
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Manuel F López-Aranda
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| | - María D López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA Plataforma Bionand, Málaga, Spain
| |
Collapse
|
4
|
Li J, Liu S, Chen C, Deng J, Du Z, Yang S, Deng H, Zhang Z, Huang Y, Fu J, Zhang W, Poon WS, Hou H, Wang J. The Epileptiogenic Modified Therapy: Regulating the Dynamic of Microglia via ROS-Responsive Cascade Nano-Formulation. Adv Healthc Mater 2024:e2403700. [PMID: 39713896 DOI: 10.1002/adhm.202403700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/13/2024] [Indexed: 12/24/2024]
Abstract
Pharmacological treatment of epilepsy presents several challenges, particularly the ineffectiveness of antiseizure medicines (ASMs) in modifying disease. In fact, the removal of reactive oxygen species (ROS) and preconditioning with tolerable dose of nitric oxide (NO) can activate neuroprotective mechanisms during latency and enhance tolerance to oxidative stress during seizures. To address this, a ROS-responsive cascade Nano-formulation (RRCN) is developed, which will transform ROS into NO. Remarkably, RRCN significantly reduces seizure severity, prolongs seizure latency, and extends inter-seizure intervals, though it does not increase the latency of generalized seizures. Microglia, the primary immune cells of the brain, play a crucial role in the initiation and progression of epilepsy. In the kainic acid (KA) epilepsy model, microglial processes elongate, branching increases, and interactions between microglia and neurons are strengthened in the CA1 and CA3 regions of the hippocampus compared to the Vehicle group. RRCN reverses these dynamic changes in microglia and their interactions with neurons, which are mediated by the NO/HIF/ErBb2 pathway. Thus, RRCN can inhibit seizures generalization by preconditioning the dynamic changes of microglia.
Collapse
Affiliation(s)
- Jiaxin Li
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chenghan Chen
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
- Department of Surgery, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Jiahong Deng
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
- Department of Surgery, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Zibo Du
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Simin Yang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Hongying Deng
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Zhixia Zhang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
- Department of Surgery, Nanfang Hospital Southern Medical University, Guangzhou, 510515, China
| | - Yiyu Huang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Jingwen Fu
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Wangming Zhang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| | - Wai Sang Poon
- Department of Surgery, Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jun Wang
- Department of Neurosurgery, The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
5
|
Stables J, Pal R, Bradford BM, Carter-Cusack D, Taylor I, Pridans C, Khan N, Woodruff TM, Irvine KM, Summers KM, Mabbott NA, Hume DA. The effect of a dominant kinase-dead Csf1r mutation associated with adult-onset leukoencephalopathy on brain development and neuropathology. Neurobiol Dis 2024; 203:106743. [PMID: 39581554 DOI: 10.1016/j.nbd.2024.106743] [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: 10/14/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Amino acid substitutions in the kinase domain of the human CSF1R protein are associated with autosomal dominant adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). To model the human disease, we created a disease-associated mutation (Glu631Lys; E631K) in the mouse Csf1r locus. Previous analysis demonstrated that heterozygous mutation (Csf1rE631K/+) had a dominant inhibitory effect on CSF1R signaling in vitro and in vivo but did not recapitulate human disease pathology. We speculated that leukoencephalopathy in humans requires an environmental trigger and/or epistatic interaction with common neurodegenerative disease-associated alleles. Here we examine the Csf1rE631K/+ mutation impact on microglial phenotype, postnatal brain development, age-related changes in gene expression and on prion disease and experimental autoimmune encephalitis (EAE), two pathologies in which microgliosis is a prominent feature. The Csf1rE631K/+ mutation reduced microglial abundance and the expression of microglial-associated transcripts relative to wild-type controls at 12 and 43 weeks of age. There was no selective effect on homeostatic markers e.g. P2ry12, or age-related changes in gene expression in striatum and hippocampus. An epistatic interaction was demonstrated between Csf1rE631K/+ and Cx3cr1EGFP/+ genotypes leading to dysregulated microglial and neuronal gene expression in hippocampus and striatum. Heterozygous Csf1rE631K mutation reduced the microgliosis associated with both diseases. There was no significant impact on disease severity or progression in prion disease. In EAE, inflammation-associated transcripts in the hippocampus and striatum were suppressed in parallel with microglia-specific transcripts. The results support a dominant inhibitory model of CSF1R-related leukoencephalopathy and likely contributions of an environmental trigger and/or genetic background to neuropathology.
Collapse
Affiliation(s)
- Jennifer Stables
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia; Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Reiss Pal
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Barry M Bradford
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Dylan Carter-Cusack
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Isis Taylor
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Clare Pridans
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Nemat Khan
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Katharine M Irvine
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Neil A Mabbott
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
6
|
Ayoub R, Yang S, Ji H, Fan L, De Michino S, Mabbott DJ, Nieman BJ. Brain volume and microglial density changes are correlated in a juvenile mouse model of cranial radiation and CSF1R inhibitor treatment. NMR IN BIOMEDICINE 2024; 37:e5222. [PMID: 39164196 DOI: 10.1002/nbm.5222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 05/30/2024] [Accepted: 06/27/2024] [Indexed: 08/22/2024]
Abstract
Microglia have been shown to proliferate and become activated following cranial radiotherapy (CRT), resulting in a chronic inflammatory response. We investigated the role of microglia in contributing to widespread volume losses observed in the brain following CRT in juvenile mice. To manipulate microglia, we used low-dose treatment with a highly selective CSF1R inhibitor called PLX5622 (PLX). We hypothesized that alteration of the post-CRT microglia population would lead to changes in brain development outcomes, as evaluated by structural MRI. Wild-type C57BL/6J mice were provided with daily intraperitoneal injections of PLX (25 mg/kg) or vehicle from postnatal day (P)14 to P19. Mice also received whole-brain irradiation (7 Gy) or sham irradiation (0 Gy) at 16 days of age. In one cohort of mice, immunohistochemical assessment in tissue sections was conducted to assess the impact of the selected PLX and CRT doses as well as their combination. In a separate cohort, mice were imaged using MRI at P14 (pretreatment), P19, P23, P42 and P63 in order to assess induced volume changes, which were measured based on structures from a predefined atlas. We observed that PLX and radiation treatments led to sex-specific changes in the microglial cell population. Across treatment groups, MRI-detected anatomical volumes at P19 and P63 were associated with microglia and proliferating microglia densities, respectively. Overall, our study demonstrates that low-dose PLX treatment produces a sex-dependent response in juvenile mice, that manipulation of microglia alters CRT-induced volume changes and that microglia density and MRI-derived volume changes are correlated in this model.
Collapse
Affiliation(s)
- Ramy Ayoub
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sabrina Yang
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Helen Ji
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lloyd Fan
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Steven De Michino
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Donald J Mabbott
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brian J Nieman
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Tisi A, Scipioni L, Carozza G, Di Re L, Cimino G, Di Meo C, Palaniappan S, Valle FD, Fanti F, Giacovazzo G, Compagnone D, Maccarone R, Oddi S, Maccarrone M. Alterations of endocannabinoid signaling and microglia reactivity in the retinas of AD-like mice precede the onset of hippocampal β-amyloid plaques. J Neurochem 2024. [PMID: 39556462 DOI: 10.1111/jnc.16256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024]
Abstract
Extra-cerebral manifestations of Alzheimer's disease (AD) develop in the retina, which is, therefore, considered a "window to the brain". Recent studies demonstrated the dysregulation of the endocannabinoid (eCB) system (ECS) in AD brain. Here, we explored the possible alterations of ECS and the onset of gliosis in the retina of AD-like mice. Tg2576 (TG) mice overexpressing the amyloid precursor protein (APP) were used at the age of 12 months, when hippocampal β-amyloid plaques had not been developed yet. Analysis of retinal gliosis showed a significant increase in the number of IBA1 (+) microglia cells in TG versus wild type (WT). Gliosis was not associated with retinal β-amyloid plaques, evident retinal degenerative signatures, or excitotoxicity; instead, oxidative stress burden was observed as increased acrolein levels. Analysis of the ECS (receptors/metabolic enzymes) through western blotting (WB) revealed the up-regulation of cannabinoid receptor 2 (CB2) and monoacylglycerol lipase (MAGL), the enzyme responsible for the degradation of 2-arachidonoylglycerol (2-AG), in TG retinas. Fluorescence intensity analysis of anti-CB2 and anti-MAGL immuno-stained cryosections was consistent with WB, showing their up-regulation throughout the retinal layers. No statistically significant differences were found for the other enzymes/receptors of the ECS under study. However, linear regression analysis for individual animals showed a significant correlation between CB2 and fatty acid amide hydrolase (FAAH), diacylglycerol lipase α/β (DAGLα/β), and APP; instead, a significant negative correlation was found between MAGL and APP. Finally, ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) demonstrated a significant reduction of 2-AG in TG retinas (~0.34 ng/mg) compared to WT (~1.70 ng/mg), while a trend toward increase was found for the other eCB anandamide (AEA). Overall, our data indicate that gliosis and ECS dysregulation-in particular of CB2, MAGL and 2-AG-occur in the retina of AD-like mice before retinal degeneration and development of hippocampal β-amyloid plaques.
Collapse
Affiliation(s)
- Annamaria Tisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Lucia Scipioni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
- Laboratory of Lipid Neurochemistry, European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, Rome, Italy
| | - Giulia Carozza
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Lucia Di Re
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giacomo Cimino
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Camilla Di Meo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Sakthimala Palaniappan
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesco Della Valle
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Federico Fanti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Giacomo Giacovazzo
- Laboratory of Lipid Neurochemistry, European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Dario Compagnone
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Rita Maccarone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Sergio Oddi
- Laboratory of Lipid Neurochemistry, European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
- Laboratory of Lipid Neurochemistry, European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, Rome, Italy
| |
Collapse
|
8
|
Zong T, Li N, Han F, Liu J, Deng M, Li V, Zhang M, Zhou Y, Yu M. Microglial depletion rescues spatial memory impairment caused by LPS administration in adult mice. PeerJ 2024; 12:e18552. [PMID: 39559328 PMCID: PMC11572354 DOI: 10.7717/peerj.18552] [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: 08/13/2024] [Accepted: 10/29/2024] [Indexed: 11/20/2024] Open
Abstract
Recent studies have highlighted the importance of microglia, the resident macrophages in the brain, in regulating cognitive functions such as learning and memory in both healthy and diseased states. However, there are conflicting results and the underlying mechanisms are not fully understood. In this study, we examined the effect of depleting adult microglia on spatial learning and memory under both physiological conditions and lipopolysaccharide (LPS)-induced neuroinflammation. Our results revealed that microglial depletion by PLX5622 caused mild spatial memory impairment in mice under physiological conditions; however, it prevented memory deficits induced by systemic LPS insult. Inactivating microglia through minocycline administration replicated the protective effect of microglial depletion on LPS-induced memory impairment. Furthermore, our study showed that PLX5622 treatment suppressed LPS-induced neuroinflammation, microglial activation, and synaptic dysfunction. These results strengthen the evidence for the involvement of microglial immunoactivation in LPS-induced synaptic and cognitive malfunctions. They also suggest that targeting microglia may be a potential approach to treating neuroinflammation-associated cognitive dysfunction seen in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tao Zong
- Affiliated Qingdao Third People’s Hospital, Department of Otorhinolaryngology Head and Neck, Qingdao University, Qingdao, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
| | - Na Li
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Qingdao Binhai University, Qingdao, Shandong, China
| | - Fubing Han
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China, China
| | - Junru Liu
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, China, China
| | - Mingru Deng
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, China, China
| | - Vincent Li
- Beverly Hills High School, Unaffiliated, Beverly Hills, California, United States
| | - Meng Zhang
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
| | - Yu Zhou
- Affiliated Qingdao Third People’s Hospital, Department of Otorhinolaryngology Head and Neck, Qingdao University, Qingdao, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, China, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Ming Yu
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
9
|
Song Q, E S, Zhang Z, Liang Y. Neuroplasticity in the transition from acute to chronic pain. Neurotherapeutics 2024; 21:e00464. [PMID: 39438166 PMCID: PMC11585895 DOI: 10.1016/j.neurot.2024.e00464] [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: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Acute pain is a transient sensation that typically serves as part of the body's defense mechanism. However, in certain patients, acute pain can evolve into chronic pain, which persists for months or even longer. Neuroplasticity refers to the capacity for variation and adaptive alterations in the morphology and functionality of neurons and synapses, and it plays a significant role in the transmission and modulation of pain. In this paper, we explore the molecular mechanisms and signaling pathways underlying neuroplasticity during the transition of pain. We also examine the effects of neurotransmitters, inflammatory mediators, and central sensitization on neuroplasticity, as well as the potential of neuroplasticity as a therapeutic strategy for preventing chronic pain. The aims of this article is to clarify the role of neuroplasticity in the transformation from acute pain to chronic pain, with the hope of providing a novel theoretical basis for unraveling the pathogenesis of chronic pain and offering more effective strategies and approaches for its diagnosis and treatment.
Collapse
Affiliation(s)
- Qingbiao Song
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China
| | - Sihan E
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China
| | - Zhiyu Zhang
- Department of Orthopedics, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, China
| | - Yingxia Liang
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
10
|
Heuer SE, Bloss EB, Howell GR. Strategies to dissect microglia-synaptic interactions during aging and in Alzheimer's disease. Neuropharmacology 2024; 254:109987. [PMID: 38705570 DOI: 10.1016/j.neuropharm.2024.109987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Age is the largest risk factor for developing Alzheimer's disease (AD), a neurodegenerative disorder that causes a progressive and severe dementia. The underlying cause of cognitive deficits seen in AD is thought to be the disconnection of neural circuits that control memory and executive functions. Insight into the mechanisms by which AD diverges from normal aging will require identifying precisely which cellular events are driven by aging and which are impacted by AD-related pathologies. Since microglia, the brain-resident macrophages, are known to have critical roles in the formation and maintenance of neural circuits through synaptic pruning, they are well-positioned to modulate synaptic connectivity in circuits sensitive to aging or AD. In this review, we provide an overview of the current state of the field and on emerging technologies being employed to elucidate microglia-synaptic interactions in aging and AD. We also discuss the importance of leveraging genetic diversity to study how these interactions are shaped across more realistic contexts. We propose that these approaches will be essential to define specific aging- and disease-relevant trajectories for more personalized therapeutics aimed at reducing the effects of age or AD pathologies on the brain. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Sarah E Heuer
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Erik B Bloss
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, USA.
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, 04469, USA.
| |
Collapse
|
11
|
Sierra A, Miron VE, Paolicelli RC, Ransohoff RM. Microglia in Health and Diseases: Integrative Hubs of the Central Nervous System (CNS). Cold Spring Harb Perspect Biol 2024; 16:a041366. [PMID: 38438189 PMCID: PMC11293550 DOI: 10.1101/cshperspect.a041366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Microglia are usually referred to as "the innate immune cells of the brain," "the resident macrophages of the central nervous system" (CNS), or "CNS parenchymal macrophages." These labels allude to their inherent immune function, related to their macrophage lineage. However, beyond their classic innate immune responses, microglia also play physiological roles crucial for proper brain development and maintenance of adult brain homeostasis. Microglia sense both external and local stimuli through a variety of surface receptors. Thus, they might serve as integrative hubs at the interface between the external environment and the CNS, able to decode, filter, and buffer cues from outside, with the aim of preserving and maintaining brain homeostasis. In this perspective, we will cast a critical look at how these multiple microglial functions are acquired and coordinated, and we will speculate on their impact on human brain physiology and pathology.
Collapse
Affiliation(s)
- Amanda Sierra
- Achucarro Basque Center for Neuroscience, Glial Cell Biology Laboratory, Science Park of UPV/EHU, E-48940 Leioa, Bizkaia, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country EHU/UPV, 48940 Leioa, Spain
- Ikerbasque Foundation, Bilbao 48009, Spain
| | - Veronique E Miron
- BARLO Multiple Sclerosis Centre, Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto M5B 1T8, Canada
- Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4TJ, United Kingdom
| | - Rosa C Paolicelli
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, CH-1005 Lausanne, Switzerland
| | | |
Collapse
|
12
|
Wickel J, Chung HY, Ceanga M, von Stackelberg N, Hahn N, Candemir Ö, Baade-Büttner C, Mein N, Tomasini P, Woldeyesus DM, Andreas N, Baumgarten P, Koch P, Groth M, Wang ZQ, Geis C. Repopulated microglia after pharmacological depletion decrease dendritic spine density in adult mouse brain. Glia 2024; 72:1484-1500. [PMID: 38780213 DOI: 10.1002/glia.24541] [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: 06/15/2023] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
Microglia are innate immune cells in the brain and show exceptional heterogeneity. They are key players in brain physiological development regulating synaptic plasticity and shaping neuronal networks. In pathological disease states, microglia-induced synaptic pruning mediates synaptic loss and targeting microglia was proposed as a promising therapeutic strategy. However, the effect of microglia depletion and subsequent repopulation on dendritic spine density and neuronal function in the adult brain is largely unknown. In this study, we investigated whether pharmacological microglia depletion affects dendritic spine density after long-term permanent microglia depletion and after short-term microglia depletion with subsequent repopulation. Long-term microglia depletion using colony-stimulating-factor-1 receptor (CSF1-R) inhibitor PLX5622 resulted in increased overall spine density, especially of mushroom spines, and increased excitatory postsynaptic current amplitudes. Short-term PLX5622 treatment with subsequent repopulation of microglia had an opposite effect resulting in activated microglia with increased synaptic phagocytosis and consequently decreased spine density and reduced excitatory neurotransmission, while Barnes maze and elevated plus maze testing was unaffected. Moreover, RNA sequencing data of isolated repopulated microglia showed an activated and proinflammatory phenotype. Long-term microglia depletion might be a promising therapeutic strategy in neurological diseases with pathological microglial activation, synaptic pruning, and synapse loss. However, repopulation after depletion induces activated microglia and results in a decrease of dendritic spines possibly limiting the therapeutic application of microglia depletion. Instead, persistent modulation of pathological microglia activity might be beneficial in controlling synaptic damage.
Collapse
Affiliation(s)
- Jonathan Wickel
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Ha-Yeun Chung
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Mihai Ceanga
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Nikolai von Stackelberg
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Nina Hahn
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Özge Candemir
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Carolin Baade-Büttner
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Nils Mein
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Paula Tomasini
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Dan M Woldeyesus
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Nico Andreas
- Department of Neurosurgery, Jena University Hospital, Jena, Germany
| | - Peter Baumgarten
- Department of Neurosurgery, Jena University Hospital, Jena, Germany
| | - Philipp Koch
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Marco Groth
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
- Faculty of Biological Sciences, Friedrich-Schiller-University, Jena, Germany
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Christian Geis
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| |
Collapse
|
13
|
Malik S, Xavier S, Soch A, Younesi S, Yip J, Slayo M, Barrientos RM, Sominsky L, Spencer SJ. High-fat diet and aging-associated memory impairments persist in the absence of microglia in female rats. Neurobiol Aging 2024; 140:22-32. [PMID: 38703636 DOI: 10.1016/j.neurobiolaging.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
Aging is associated with a priming of microglia such that they are hypersensitive to further immune challenges. As such high-fat diet during aging can have detrimental effects on cognition that is not seen in the young. However, conflicting findings also suggest that obesity may protect against cognitive decline during aging. Given this uncertainty we aimed here to examine the role of microglia in high-fat, high-sucrose diet (HFSD)-induced changes in cognitive performance in the aging brain. We hypothesised that 8 weeks of HFSD-feeding would alter microglia and the inflammatory milieu in aging and worsen aging-related cognitive deficits in a microglia-dependent manner. We found that both aging and HFSD reduced hippocampal neuron numbers and open field exploration; they also impaired recognition memory. However, the aging-related deficits occurred in the absence of a pro-inflammatory response and the deficits in memory performance persisted after depletion of microglia in the Cx3cr1-Dtr knock-in rat. Our data suggest that mechanisms additional to the acute microglial contribution play a role in aging- and HFSD-associated memory dysfunction.
Collapse
Affiliation(s)
- Sajida Malik
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Soniya Xavier
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia
| | - Alita Soch
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Simin Younesi
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jackson Yip
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia
| | - Mary Slayo
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Germany; Center for Mind, Brain and Behavior-CMBB, Giessen, Marburg, Germany
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| | - Luba Sominsky
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia; Barwon Health, Geelong, Victoria, Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Berki P, Cserép C, Környei Z, Pósfai B, Szabadits E, Domonkos A, Kellermayer A, Nyerges M, Wei X, Mody I, Kunihiko A, Beck H, Kaikai H, Ya W, Lénárt N, Wu Z, Jing M, Li Y, Gulyás AI, Dénes Á. Microglia contribute to neuronal synchrony despite endogenous ATP-related phenotypic transformation in acute mouse brain slices. Nat Commun 2024; 15:5402. [PMID: 38926390 PMCID: PMC11208608 DOI: 10.1038/s41467-024-49773-1] [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: 12/30/2023] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Acute brain slices represent a workhorse model for studying the central nervous system (CNS) from nanoscale events to complex circuits. While slice preparation inherently involves tissue damage, it is unclear how microglia, the main immune cells and damage sensors of the CNS react to this injury and shape neuronal activity ex vivo. To this end, we investigated microglial phenotypes and contribution to network organization and functioning in acute brain slices. We reveal time-dependent microglial phenotype changes influenced by complex extracellular ATP dynamics through P2Y12R and CX3CR1 signalling, which is sustained for hours in ex vivo mouse brain slices. Downregulation of P2Y12R and changes of microglia-neuron interactions occur in line with alterations in the number of excitatory and inhibitory synapses over time. Importantly, functional microglia modulate synapse sprouting, while microglial dysfunction results in markedly impaired ripple activity both ex vivo and in vivo. Collectively, our data suggest that microglia are modulators of complex neuronal networks with important roles to maintain neuronal network integrity and activity. We suggest that slice preparation can be used to model time-dependent changes of microglia-neuron interactions to reveal how microglia shape neuronal circuits in physiological and pathological conditions.
Collapse
Affiliation(s)
- Péter Berki
- János Szentágothai Doctoral School of Neuroscience, Semmelweis University, Budapest, H-1083, Hungary
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Neuronal Network and Behaviour, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zsuzsanna Környei
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Eszter Szabadits
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Andor Domonkos
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Thalamus Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Anna Kellermayer
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Miklós Nyerges
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Xiaofei Wei
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Istvan Mody
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Araki Kunihiko
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - Heinz Beck
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - He Kaikai
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Wang Ya
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Miao Jing
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Attila I Gulyás
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary.
| |
Collapse
|
15
|
Yu A, Tan LX, Lakkaraju A, Santina LD, Ou Y. Microglia target synaptic sites early during excitatory circuit disassembly in neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598914. [PMID: 38915631 PMCID: PMC11195198 DOI: 10.1101/2024.06.13.598914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
During development, microglia prune excess synapses to refine neuronal circuits. In neurodegeneration, the role of microglia-mediated synaptic pruning in circuit remodeling and dysfunction is important for developing therapies aimed at modulating microglial function. Here we analyzed the role of microglia in the synapse disassembly of degenerating postsynaptic neurons in the inner retina. After inducing transient intraocular pressure elevation to injure retinal ganglion cells, microglia increase in number, shift to ameboid morphology, and exhibit greater process movement. Furthermore, due to the greater number of microglia, there is increased colocalization of microglia with synaptic components throughout the inner plexiform layer and with excitatory synaptic sites along individual ganglion cell dendrites. Microglia depletion partially restores ganglion cell function, suggesting that microglia activation may be neurotoxic in early neurodegeneration. Our results demonstrate the important role of microglia in synapse disassembly in degenerating circuits, highlighting their recruitment to synaptic sites early after neuronal injury.
Collapse
Affiliation(s)
- Alfred Yu
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
| | - Li Xuan Tan
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
| | - Aparna Lakkaraju
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
| | - Luca Della Santina
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
- College of Optometry, University of Houston, Houston, TX, USA
| | - Yvonne Ou
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
| |
Collapse
|
16
|
Surala M, Soso-Zdravkovic L, Munro D, Rifat A, Ouk K, Vida I, Priller J, Madry C. Lifelong absence of microglia alters hippocampal glutamatergic networks but not synapse and spine density. EMBO Rep 2024; 25:2348-2374. [PMID: 38589666 PMCID: PMC11094096 DOI: 10.1038/s44319-024-00130-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Microglia sculpt developing neural circuits by eliminating excess synapses in a process called synaptic pruning, by removing apoptotic neurons, and by promoting neuronal survival. To elucidate the role of microglia during embryonic and postnatal brain development, we used a mouse model deficient in microglia throughout life by deletion of the fms-intronic regulatory element (FIRE) in the Csf1r locus. Surprisingly, young adult Csf1rΔFIRE/ΔFIRE mice display no changes in excitatory and inhibitory synapse number and spine density of CA1 hippocampal neurons compared with Csf1r+/+ littermates. However, CA1 neurons are less excitable, receive less CA3 excitatory input and show altered synaptic properties, but this does not affect novel object recognition. Cytokine profiling indicates an anti-inflammatory state along with increases in ApoE levels and reactive astrocytes containing synaptic markers in Csf1rΔFIRE/ΔFIRE mice. Notably, these changes in Csf1rΔFIRE/ΔFIRE mice closely resemble the effects of acute microglial depletion in adult mice after normal development. Our findings suggest that microglia are not mandatory for synaptic pruning, and that in their absence pruning can be achieved by other mechanisms.
Collapse
Affiliation(s)
- Michael Surala
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Luna Soso-Zdravkovic
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany
| | - David Munro
- University of Edinburgh and UK Dementia Research Institute, Edinburgh, EH16 4TJ, UK
| | - Ali Rifat
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Koliane Ouk
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charitéplatz 1, 10117, Berlin, Germany
| | - Imre Vida
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute for Integrative Neuroanatomy, Charitéplatz 1, 10117, Berlin, Germany
| | - Josef Priller
- University of Edinburgh and UK Dementia Research Institute, Edinburgh, EH16 4TJ, UK.
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charitéplatz 1, 10117, Berlin, Germany.
- DZNE Berlin, 10117, Berlin, Germany.
- Department of Psychiatry and Psychotherapy; School of Medicine and Health, Technical University of Munich and German Center for Mental Health (DZPG), 81675, Munich, Germany.
| | - Christian Madry
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
17
|
Kim W, Kim M, Kim B. Unraveling the enigma: housekeeping gene Ugt1a7c as a universal biomarker for microglia. Front Psychiatry 2024; 15:1364201. [PMID: 38666091 PMCID: PMC11043603 DOI: 10.3389/fpsyt.2024.1364201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Background Microglia, brain resident macrophages, play multiple roles in maintaining homeostasis, including immunity, surveillance, and protecting the central nervous system through their distinct activation processes. Identifying all types of microglia-driven populations is crucial due to the presence of various phenotypes that differ based on developmental stages or activation states. During embryonic development, the E8.5 yolk sac contains erythromyeloid progenitors that go through different growth phases, eventually resulting in the formation of microglia. In addition, microglia are present in neurological diseases as a diverse population. So far, no individual biomarker for microglia has been discovered that can accurately identify and monitor their development and attributes. Summary Here, we highlight the newly defined biomarker of mouse microglia, UGT1A7C, which exhibits superior stability in expression during microglia development and activation compared to other known microglia biomarkers. The UGT1A7C sensing chemical probe labels all microglia in the 3xTG AD mouse model. The expression of Ugt1a7c is stable during development, with only a 4-fold variation, while other microglia biomarkers, such as Csf1r and Cx3cr1, exhibit at least a 10-fold difference. The UGT1A7C expression remains constant throughout its lifespan. In addition, the expression and activity of UGT1A7C are the same in response to different types of inflammatory activators' treatment in vitro. Conclusion We propose employing UGT1A7C as the representative biomarker for microglia, irrespective of their developmental state, age, or activation status. Using UGT1A7C can reduce the requirement for using multiple biomarkers, enhance the precision of microglia analysis, and even be utilized as a standard for gene/protein expression.
Collapse
Affiliation(s)
| | | | - Beomsue Kim
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| |
Collapse
|
18
|
Li Z, Yu S, Li L, Zhou C, Wang L, Tang S, Gu N, Zhang Z, Huang Z, Chen H, Tang W, Wang Y, Yang X, Sun X, Yan J. TREM2 alleviates white matter injury after traumatic brain injury in mice might be mediated by regulation of DHCR24/LXR pathway in microglia. Clin Transl Med 2024; 14:e1665. [PMID: 38649789 PMCID: PMC11035381 DOI: 10.1002/ctm2.1665] [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: 11/23/2023] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND White matter injury (WMI) is an important pathological process after traumatic brain injury (TBI). The correlation between white matter functions and the myeloid cells expressing triggering receptor-2 (TREM2) has been convincingly demonstrated. Moreover, a recent study revealed that microglial sterol metabolism is crucial for early remyelination after demyelinating diseases. However, the potential roles of TREM2 expression and microglial sterol metabolism in WMI after TBI have not yet been explored. METHODS Controlled cortical injury was induced in both wild-type (WT) and TREM2 depletion (TREM2 KO) mice to simulate clinical TBI. COG1410 was used to upregulate TREM2, while PLX5622 and GSK2033 were used to deplete microglia and inhibit the liver X receptor (LXR), respectively. Immunofluorescence, Luxol fast blue staining, magnetic resonance imaging, transmission electron microscopy, and oil red O staining were employed to assess WMI after TBI. Neurological behaviour tests and electrophysiological recordings were utilized to evaluate cognitive functions following TBI. Microglial cell sorting and transcriptomic sequencing were utilized to identify alterations in microglial sterol metabolism-related genes, while western blot was conducted to validate the findings. RESULTS TREM2 expressed highest at 3 days post-TBI and was predominantly localized to microglial cells within the white matter. Depletion of TREM2 worsened aberrant neurological behaviours, and this phenomenon was mediated by the exacerbation of WMI, reduced renewal of oligodendrocytes, and impaired phagocytosis ability of microglia after TBI. Subsequently, the upregulation of TREM2 alleviated WMI, promoted oligodendrocyte regeneration, and ultimately facilitated the recovery of neurological behaviours after TBI. Finally, the expression of DHCR24 increased in TREM2 KO mice after TBI. Interestingly, TREM2 inhibited DHCR24 and upregulated members of the LXR pathway. Moreover, LXR inhibition could partially reverse the effects of TREM2 upregulation on electrophysiological activities. CONCLUSIONS We demonstrate that TREM2 has the potential to alleviate WMI following TBI, possibly through the DHCR24/LXR pathway in microglia.
Collapse
Affiliation(s)
- Zhao Li
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Emergency DepartmentChengdu First People's HospitalChengduChina
| | - Shenghui Yu
- Emergency DepartmentChengdu First People's HospitalChengduChina
| | - Lin Li
- Department of NeurosurgeryChongqing University Cancer HospitalChongqingChina
| | - Chao Zhou
- Emergency DepartmentChengdu First People's HospitalChengduChina
| | - Lin Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of NeurosurgeryNanchong Central HospitalThe Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Shuang Tang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of NeurosurgerySuining Central HospitalSuiningChina
| | - Nina Gu
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhaosi Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhijian Huang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Hong Chen
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Wei Tang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yingwen Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaomin Yang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaochuan Sun
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jin Yan
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
19
|
Yu Z, Shi H, Zhang J, Ma C, He C, Yang F, Zhao L. ROLE OF MICROGLIA IN SEPSIS-ASSOCIATED ENCEPHALOPATHY PATHOGENESIS: AN UPDATE. Shock 2024; 61:498-508. [PMID: 38150368 DOI: 10.1097/shk.0000000000002296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
ABSTRACT Sepsis-associated encephalopathy (SAE) is a serious complication of sepsis, which is characterized by cognitive dysfunction, a poor prognosis, and high incidences of morbidity and mortality. Substantial levels of systemic inflammatory factors induce neuroinflammatory responses during sepsis, ultimately disrupting the central nervous system's (CNS) homeostasis. This disruption results in brain dysfunction through various underlying mechanisms, contributing further to SAE's development. Microglia, the most important macrophage in the CNS, can induce neuroinflammatory responses, brain tissue injury, and neuronal dysregulation, resulting in brain dysfunction. They serve an important regulatory role in CNS homeostasis and can be activated through multiple pathways. Consequently, activated microglia are involved in several pathogenic mechanisms related to SAE and play a crucial role in its development. This article discusses the role of microglia in neuroinflammation, dysfunction of neurotransmitters, disruption of the blood-brain barrier, abnormal control of cerebral blood flow, mitochondrial dysfunction, and reduction in the number of good bacteria in the gut as main pathogenic mechanisms of SAE and focuses on studies targeting microglia to ameliorate SAE to provide a theoretical basis for targeted microglial therapy for SAE.
Collapse
Affiliation(s)
| | - Hui Shi
- Department of Critical Care Medicine, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Jingjing Zhang
- Department of Central Laboratory, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Chunhan Ma
- Chifeng Clinical Medical College of Inner Mongolia Medical University, Hohhot, China
| | - Chen He
- Chifeng Clinical Medical College of Inner Mongolia Medical University, Hohhot, China
| | - Fei Yang
- Department of Critical Care Medicine, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Lina Zhao
- Department of Critical Care Medicine, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
20
|
Stowell R, Wang KH. Dopaminergic signaling regulates microglial surveillance and adolescent plasticity in the frontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584167. [PMID: 38559264 PMCID: PMC10979918 DOI: 10.1101/2024.03.08.584167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Adolescence is a sensitive period for frontal cortical development and cognitive maturation. The dopaminergic (DA) mesofrontal circuit is particularly malleable in response to changes in adolescent experience and DA activity. However, the cellular mechanisms engaged in this plasticity remain unexplored. Here, we report that microglia, the innate immune cells of the brain, are uniquely sensitive to adolescent mesofrontal DA signaling. Longitudinal in vivo two-photon imaging in mice shows that frontal cortical microglia respond dynamically to plasticity-inducing behavioral or optogenetic DA axon stimulation with increased parenchymal and DA bouton surveillance. Microglial-axon contact precedes new bouton formation, and both D1 and D2-type DA receptors regulate microglial-bouton interactions and axonal plasticity. Moreover, D2 antagonism in adults reinstates adolescent plasticity, including increased microglial surveillance and new DA bouton formation. Our results reveal that DA signaling regulates microglial surveillance and axonal plasticity uniquely in the adolescent frontal cortex, presenting potential interventions for restoring plasticity in the adult brain.
Collapse
Affiliation(s)
- Rianne Stowell
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642
| | - Kuan Hong Wang
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642
| |
Collapse
|
21
|
You Y, Chen Z, Hu WW. The role of microglia heterogeneity in synaptic plasticity and brain disorders: Will sequencing shed light on the discovery of new therapeutic targets? Pharmacol Ther 2024; 255:108606. [PMID: 38346477 DOI: 10.1016/j.pharmthera.2024.108606] [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: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Microglia play a crucial role in interacting with neuronal synapses and modulating synaptic plasticity. This function is particularly significant during postnatal development, as microglia are responsible for removing excessive synapses to prevent neurodevelopmental deficits. Dysregulation of microglial synaptic function has been well-documented in various pathological conditions, notably Alzheimer's disease and multiple sclerosis. The recent application of RNA sequencing has provided a powerful and unbiased means to decipher spatial and temporal microglial heterogeneity. By identifying microglia with varying gene expression profiles, researchers have defined multiple subgroups of microglia associated with specific pathological states, including disease-associated microglia, interferon-responsive microglia, proliferating microglia, and inflamed microglia in multiple sclerosis, among others. However, the functional roles of these distinct subgroups remain inadequately characterized. This review aims to refine our current understanding of the potential roles of heterogeneous microglia in regulating synaptic plasticity and their implications for various brain disorders, drawing from recent sequencing research and functional studies. This knowledge may aid in the identification of pathogenetic biomarkers and potential factors contributing to pathogenesis, shedding new light on the discovery of novel drug targets. The field of sequencing-based data mining is evolving toward a multi-omics approach. With advances in viral tools for precise microglial regulation and the development of brain organoid models, we are poised to elucidate the functional roles of microglial subgroups detected through sequencing analysis, ultimately identifying valuable therapeutic targets.
Collapse
Affiliation(s)
- Yi You
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
22
|
Donovan LJ, Bridges CM, Nippert AR, Wang M, Wu S, Forman TE, Haight ES, Huck NA, Bond SF, Jordan CE, Gardner AM, Nair RV, Tawfik VL. Repopulated spinal cord microglia exhibit a unique transcriptome and contribute to pain resolution. Cell Rep 2024; 43:113683. [PMID: 38261512 PMCID: PMC10947777 DOI: 10.1016/j.celrep.2024.113683] [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: 06/15/2023] [Revised: 11/15/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
Microglia are implicated as primarily detrimental in pain models; however, they exist across a continuum of states that contribute to homeostasis or pathology depending on timing and context. To clarify the specific contribution of microglia to pain progression, we take advantage of a temporally controlled transgenic approach to transiently deplete microglia. Unexpectedly, we observe complete resolution of pain coinciding with microglial repopulation rather than depletion. We find that repopulated mouse spinal cord microglia are morphologically distinct from control microglia and exhibit a unique transcriptome. Repopulated microglia from males and females express overlapping networks of genes related to phagocytosis and response to stress. We intersect the identified mouse genes with a single-nuclei microglial dataset from human spinal cord to identify human-relevant genes that may ultimately promote pain resolution after injury. This work presents a comprehensive approach to gene discovery in pain and provides datasets for the development of future microglial-targeted therapeutics.
Collapse
Affiliation(s)
- Lauren J Donovan
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Caldwell M Bridges
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Amy R Nippert
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Meng Wang
- Stanford Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Shaogen Wu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Thomas E Forman
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Elena S Haight
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Nolan A Huck
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sabrina F Bond
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Claire E Jordan
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Aysha M Gardner
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ramesh V Nair
- Stanford Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
23
|
Loh JS, Mak WQ, Tan LKS, Ng CX, Chan HH, Yeow SH, Foo JB, Ong YS, How CW, Khaw KY. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther 2024; 9:37. [PMID: 38360862 PMCID: PMC10869798 DOI: 10.1038/s41392-024-01743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/02/2024] [Accepted: 01/14/2024] [Indexed: 02/17/2024] Open
Abstract
The human gastrointestinal tract is populated with a diverse microbial community. The vast genetic and metabolic potential of the gut microbiome underpins its ubiquity in nearly every aspect of human biology, including health maintenance, development, aging, and disease. The advent of new sequencing technologies and culture-independent methods has allowed researchers to move beyond correlative studies toward mechanistic explorations to shed light on microbiome-host interactions. Evidence has unveiled the bidirectional communication between the gut microbiome and the central nervous system, referred to as the "microbiota-gut-brain axis". The microbiota-gut-brain axis represents an important regulator of glial functions, making it an actionable target to ameliorate the development and progression of neurodegenerative diseases. In this review, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases. As the gut microbiome provides essential cues to microglia, astrocytes, and oligodendrocytes, we examine the communications between gut microbiota and these glial cells during healthy states and neurodegenerative diseases. Subsequently, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases using a metabolite-centric approach, while also examining the role of gut microbiota-related neurotransmitters and gut hormones. Next, we examine the potential of targeting the intestinal barrier, blood-brain barrier, meninges, and peripheral immune system to counteract glial dysfunction in neurodegeneration. Finally, we conclude by assessing the pre-clinical and clinical evidence of probiotics, prebiotics, and fecal microbiota transplantation in neurodegenerative diseases. A thorough comprehension of the microbiota-gut-brain axis will foster the development of effective therapeutic interventions for the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jian Sheng Loh
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Wen Qi Mak
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Li Kar Stella Tan
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Chu Xin Ng
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Hong Hao Chan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Shiau Hueh Yeow
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Yong Sze Ong
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| | - Kooi Yeong Khaw
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
24
|
Kim AB, Xiao Q, Yan P, Pan Q, Pandey G, Grathwohl S, Gonzales E, Xu I, Cho Y, Haecker H, Epelman S, Diwan A, Lee JM, DeSelm CJ. Chimeric antigen receptor macrophages target and resorb amyloid plaques. JCI Insight 2024; 9:e175015. [PMID: 38516884 PMCID: PMC11063938 DOI: 10.1172/jci.insight.175015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Substantial evidence suggests a role for immunotherapy in treating Alzheimer's disease (AD). While the precise pathophysiology of AD is incompletely understood, clinical trials of antibodies targeting aggregated forms of β amyloid (Aβ) have shown that reducing amyloid plaques can mitigate cognitive decline in patients with early-stage AD. Here, we describe what we believe to be a novel approach to target and degrade amyloid plaques by genetically engineering macrophages to express an Aβ-targeting chimeric antigen receptor (CAR-Ms). When injected intrahippocampally, first-generation CAR-Ms have limited persistence and fail to significantly reduce plaque load, which led us to engineer next-generation CAR-Ms that secrete M-CSF and self-maintain without exogenous cytokines. Cytokine secreting "reinforced CAR-Ms" have greater survival in the brain niche and significantly reduce plaque load locally in vivo. These findings support CAR-Ms as a platform to rationally target, resorb, and degrade pathogenic material that accumulates with age, as exemplified by targeting Aβ in AD.
Collapse
Affiliation(s)
- Alexander B. Kim
- Department of Radiation Oncology
- Bursky Center for Human Immunology and Immunotherapy
| | - Qingli Xiao
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ping Yan
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qiuyun Pan
- Department of Radiation Oncology
- Bursky Center for Human Immunology and Immunotherapy
| | - Gaurav Pandey
- Department of Radiation Oncology
- Bursky Center for Human Immunology and Immunotherapy
| | - Susie Grathwohl
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ernesto Gonzales
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Isabella Xu
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yoonho Cho
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hans Haecker
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Slava Epelman
- Department of Medicine, Division of Cardiology, Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Abhinav Diwan
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
- Departments of Medicine, Cell Biology and Physiology, Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
- Medicine Service, St. Louis VA Medical Center, St. Louis, Missouri, USA
| | - Jin-Moo Lee
- Department of Neurology, and
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carl J. DeSelm
- Department of Radiation Oncology
- Bursky Center for Human Immunology and Immunotherapy
| |
Collapse
|
25
|
Salvi J, Andreoletti P, Audinat E, Balland E, Ben Fradj S, Cherkaoui-Malki M, Heurtaux T, Liénard F, Nédélec E, Rovère C, Savary S, Véjux A, Trompier D, Benani A. Microgliosis: a double-edged sword in the control of food intake. FEBS J 2024; 291:615-631. [PMID: 35880408 DOI: 10.1111/febs.16583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 02/16/2024]
Abstract
Maintaining energy balance is essential for survival and health. This physiological function is controlled by the brain, which adapts food intake to energy needs. Indeed, the brain constantly receives a multitude of biological signals that are derived from digested foods or that originate from the gastrointestinal tract, energy stores (liver and adipose tissues) and other metabolically active organs (muscles). These signals, which include circulating nutrients, hormones and neuronal inputs from the periphery, collectively provide information on the overall energy status of the body. In the brain, several neuronal populations can specifically detect these signals. Nutrient-sensing neurons are found in discrete brain areas and are highly enriched in the hypothalamus. In turn, specialized brain circuits coordinate homeostatic responses acting mainly on appetite, peripheral metabolism, activity and arousal. Accumulating evidence shows that hypothalamic microglial cells located at the vicinity of these circuits can influence the brain control of energy balance. However, microglial cells could have opposite effects on energy balance, that is homeostatic or detrimental, and the conditions for this shift are not totally understood yet. One hypothesis relies on the extent of microglial activation, and nutritional lipids can considerably change it.
Collapse
Affiliation(s)
- Juliette Salvi
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Etienne Audinat
- IGF, Université de Montpellier, CNRS, Inserm, Montpellier, France
| | - Eglantine Balland
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Notting Hill, Australia
| | - Selma Ben Fradj
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | | | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Fabienne Liénard
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nédélec
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Carole Rovère
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | - Stéphane Savary
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Anne Véjux
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Doriane Trompier
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
26
|
Balestri W, Sharma R, da Silva VA, Bobotis BC, Curle AJ, Kothakota V, Kalantarnia F, Hangad MV, Hoorfar M, Jones JL, Tremblay MÈ, El-Jawhari JJ, Willerth SM, Reinwald Y. Modeling the neuroimmune system in Alzheimer's and Parkinson's diseases. J Neuroinflammation 2024; 21:32. [PMID: 38263227 PMCID: PMC10807115 DOI: 10.1186/s12974-024-03024-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/16/2024] [Indexed: 01/25/2024] Open
Abstract
Parkinson's disease (PD) and Alzheimer's disease (AD) are neurodegenerative disorders caused by the interaction of genetic, environmental, and familial factors. These diseases have distinct pathologies and symptoms that are linked to specific cell populations in the brain. Notably, the immune system has been implicated in both diseases, with a particular focus on the dysfunction of microglia, the brain's resident immune cells, contributing to neuronal loss and exacerbating symptoms. Researchers use models of the neuroimmune system to gain a deeper understanding of the physiological and biological aspects of these neurodegenerative diseases and how they progress. Several in vitro and in vivo models, including 2D cultures and animal models, have been utilized. Recently, advancements have been made in optimizing these existing models and developing 3D models and organ-on-a-chip systems, holding tremendous promise in accurately mimicking the intricate intracellular environment. As a result, these models represent a crucial breakthrough in the transformation of current treatments for PD and AD by offering potential for conducting long-term disease-based modeling for therapeutic testing, reducing reliance on animal models, and significantly improving cell viability compared to conventional 2D models. The application of 3D and organ-on-a-chip models in neurodegenerative disease research marks a prosperous step forward, providing a more realistic representation of the complex interactions within the neuroimmune system. Ultimately, these refined models of the neuroimmune system aim to aid in the quest to combat and mitigate the impact of debilitating neuroimmune diseases on patients and their families.
Collapse
Affiliation(s)
- Wendy Balestri
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham, UK
- Medical Technologies Innovation Facility, Nottingham Trent University, Nottingham, UK
| | - Ruchi Sharma
- Department of Mechanical Engineering, University of Victoria, Victoria, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Victor A da Silva
- Department of Mechanical Engineering, University of Victoria, Victoria, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Bianca C Bobotis
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Annabel J Curle
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Vandana Kothakota
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | | | - Maria V Hangad
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Department of Chemistry, University of Victoria, Victoria, BC, Canada
| | - Mina Hoorfar
- Department of Mechanical Engineering, University of Victoria, Victoria, Canada
| | - Joanne L Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Neurosciences Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Institute On Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Jehan J El-Jawhari
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Stephanie M Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| | - Yvonne Reinwald
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham, UK.
- Medical Technologies Innovation Facility, Nottingham Trent University, Nottingham, UK.
| |
Collapse
|
27
|
Weyer MP, Strehle J, Schäfer MKE, Tegeder I. Repurposing of pexidartinib for microglia depletion and renewal. Pharmacol Ther 2024; 253:108565. [PMID: 38052308 DOI: 10.1016/j.pharmthera.2023.108565] [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: 09/28/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Pexidartinib (PLX3397) is a small molecule receptor tyrosine kinase inhibitor of colony stimulating factor 1 receptor (CSF1R) with moderate selectivity over other members of the platelet derived growth factor receptor family. It is approved for treatment of tenosynovial giant cell tumors (TGCT). CSF1R is highly expressed by microglia, which are macrophages of the central nervous system (CNS) that defend the CNS against injury and pathogens and contribute to synapse development and plasticity. Challenged by pathogens, apoptotic cells, debris, or inflammatory molecules they adopt a responsive state to propagate the inflammation and eventually return to a homeostatic state. The phenotypic switch may fail, and disease-associated microglia contribute to the pathophysiology in neurodegenerative or neuropsychiatric diseases or long-lasting detrimental brain inflammation after brain, spinal cord or nerve injury or ischemia/hemorrhage. Microglia also contribute to the growth permissive tumor microenvironment of glioblastoma (GBM). In rodents, continuous treatment for 1-2 weeks via pexidartinib food pellets leads to a depletion of microglia and subsequent repopulation from the remaining fraction, which is aided by peripheral monocytes that search empty niches for engraftment. The putative therapeutic benefit of such microglia depletion or forced renewal has been assessed in almost any rodent model of CNS disease or injury or GBM with heterogeneous outcomes, but a tendency of partial beneficial effects. So far, microglia monitoring e.g. via positron emission imaging is not standard of care for patients receiving Pexidartinib (e.g. for TGCT), so that the depletion and repopulation efficiency in humans is still largely unknown. Considering the virtuous functions of microglia, continuous depletion is likely no therapeutic option but short-lasting transient partial depletion to stimulate microglia renewal or replace microglia in genetic disease in combination with e.g. stem cell transplantation or as part of a multimodal concept in treatment of glioblastoma appears feasible. The present review provides an overview of the preclinical evidence pro and contra microglia depletion as a therapeutic approach.
Collapse
Affiliation(s)
- Marc-Philipp Weyer
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany
| | - Jenny Strehle
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany.
| |
Collapse
|
28
|
Reverte I, Marchetti C, Pezza S, Zenoni SF, Scaringi G, Ferrucci L, D'Ottavio G, Pignataro A, Andolina D, Raspa M, Scavizzi F, Venniro M, Ramsey LA, Gross C, Caprioli D, Ragozzino D. Microglia-mediated calcium-permeable AMPAR accumulation in the nucleus accumbens drives hyperlocomotion during cocaine withdrawal. Brain Behav Immun 2024; 115:535-542. [PMID: 37967660 PMCID: PMC10915906 DOI: 10.1016/j.bbi.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/19/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023] Open
Abstract
During withdrawal from cocaine, calcium permeable-AMPA receptors (CP-AMPAR) progressively accumulate in nucleus accumbens (NAc) synapses, a phenomenon linked to behavioral sensitization and drug-seeking. Recently, it has been suggested that neuroimmune alterations might promote aberrant changes in synaptic plasticity, thus contributing to substance abuse-related behaviors. Here, we investigated the role of microglia in NAc neuroadaptations after withdrawal from cocaine-induced conditioned place preference (CPP). We depleted microglia using PLX5622-supplemented diet during cocaine withdrawal, and after the place preference test, we measured dendritic spine density and the presence of CP-AMPAR in the NAc shell. Microglia depletion prevented cocaine-induced changes in dendritic spines and CP-AMPAR accumulation. Furthermore, microglia depletion prevented conditioned hyperlocomotion without affecting drug-context associative memory. Microglia displayed fewer number of branches, resulting in a reduced arborization area and microglia control domain at late withdrawal. Our results suggest that microglia are necessary for the synaptic adaptations in NAc synapses during cocaine withdrawal and therefore represent a promising therapeutic target for relapse prevention.
Collapse
Affiliation(s)
- Ingrid Reverte
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Claudia Marchetti
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Sara Pezza
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Soami F Zenoni
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giorgia Scaringi
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Laura Ferrucci
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Ginevra D'Ottavio
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Annabella Pignataro
- IRCCS Santa Lucia Foundation, Rome, Italy; Institute of Translational Pharmacology, National Research Council, CNR, Rome, Italy
| | - Diego Andolina
- IRCCS Santa Lucia Foundation, Rome, Italy; Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Marcello Raspa
- National Research Council, Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), International Campus "A. Buzzati-Traverso", Monterotondo (Rome), Italy
| | - Ferdinando Scavizzi
- National Research Council, Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), International Campus "A. Buzzati-Traverso", Monterotondo (Rome), Italy
| | - Marco Venniro
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, USA
| | - Leslie A Ramsey
- Behavioral Neuroscience Research Branch, Intramural Research Program, Baltimore NIDA, NIH, USA
| | - Cornelius Gross
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory (EMBL), Monterotondo, Italy
| | - Daniele Caprioli
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy.
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Institute Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
29
|
Vecchiarelli HA, Lopes LT, Paolicelli RC, Stevens B, Wake H, Tremblay MÈ. Synapse Regulation. ADVANCES IN NEUROBIOLOGY 2024; 37:179-208. [PMID: 39207693 DOI: 10.1007/978-3-031-55529-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are the resident immune cells of the brain. As such, they rapidly detect changes in normal brain homeostasis and accurately respond by fine-tuning in a tightly regulated manner their morphology, gene expression, and functional behavior. Depending on the nature of these changes, microglia can thicken and retract their processes, proliferate and migrate, release numerous signaling factors and compounds influencing neuronal physiology (e.g., cytokines and trophic factors), in addition to secreting proteases able to transform the extracellular matrix, and phagocytosing various types of cellular debris, etc. Because microglia also transform rapidly (on a time scale of minutes) during experimental procedures, studying these very special cells requires methods that are specifically non-invasive. The development of such methods has provided unprecedented insights into the roles of microglia during normal physiological conditions. In particular, transcranial two-photon in vivo imaging revealed that presumably "resting" microglia continuously survey the brain parenchyma with their highly motile processes, in addition to modulating their structural and functional interactions with neuronal circuits along the changes in neuronal activity and behavioral experience occurring throughout the lifespan. In this chapter, we will describe how surveillant microglia interact with synaptic elements and modulate the number, maturation, function, and plasticity of synapses in the healthy developing, mature, and aging brain, with consequences on neuronal activity, learning and memory, and the behavioral outcome.
Collapse
Affiliation(s)
| | | | - Rosa C Paolicelli
- Division of Psychiatry Research, University of Zurich, Schlieren, Switzerland
| | - Beth Stevens
- Department of Neurology, Harvard Medical School, Center for Life Science, Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA
| | - Hiroaki Wake
- Division of Brain Circuits, National Institute for Basic Biology, Myodaiji-cho, Okazaki, Japan
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
30
|
Filho AMC, Gomes NS, Lós DB, Leite IB, Tremblay MÈ, Macêdo DS. Microglia and Microbiome-Gut-Brain Axis. ADVANCES IN NEUROBIOLOGY 2024; 37:303-331. [PMID: 39207699 DOI: 10.1007/978-3-031-55529-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The mammalian gut contains a community of microorganisms called gut microbiome. The gut microbiome is integrated into mammalian physiology, contributing to metabolism, production of metabolites, and promoting immunomodulatory actions. Microglia, the brain's resident innate immune cells, play an essential role in homeostatic neurogenesis, synaptic remodeling, and glial maturation. Microglial dysfunction has been implicated in the pathogenesis of several neuropsychiatric disorders. Recent findings indicate that microglia are influenced by the gut microbiome and their derived metabolites throughout life. The pathways by which microbiota regulate microglia have only started to be understood, but this discovery has the potential to provide valuable insights into the pathogenesis of brain disorders associated with an altered microbiome. Here, we discuss the recent literature on the role of the gut microbiome in modulating microglia during development and adulthood and summarize the key findings on this bidirectional crosstalk in selected examples of neuropsychiatric and neurodegenerative disorders. We also highlight some current caveats and perspectives for the field.
Collapse
Affiliation(s)
- Adriano Maia Chaves Filho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Nayana Soares Gomes
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Deniele Bezerra Lós
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Isabel Bessa Leite
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Department of Molecular Medicine, Université de Laval, Québec City, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Danielle S Macêdo
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
31
|
Pinto MJ, Ragozzino D, Bessis A, Audinat E. Microglial Modulation of Synaptic Maturation, Activity, and Plasticity. ADVANCES IN NEUROBIOLOGY 2024; 37:209-219. [PMID: 39207694 DOI: 10.1007/978-3-031-55529-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, which are the resident immune cells of the CNS, also have important functions in physiological conditions. In this chapter, we review the experimental evidence that microglia modulate neuronal and synaptic activity during normal development and in adults. We show that microglia can regulate the maturation and function of both inhibitory and excitatory synapses that can be stimulated or repressed. We further review the fact that these regulations occur in various brain regions, through soluble and membrane molecules, directly or through other cell partners. This review emphasizes the fact that microglia are genuine and highly context-dependent and thus adaptable regulators of neuronal activity.
Collapse
Affiliation(s)
- Maria Joana Pinto
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
- Center for Neuroscience and Cell Biology (CNC), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - Alain Bessis
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Etienne Audinat
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
32
|
Šimončičová E, Henderson Pekarik K, Vecchiarelli HA, Lauro C, Maggi L, Tremblay MÈ. Adult Neurogenesis, Learning and Memory. ADVANCES IN NEUROBIOLOGY 2024; 37:221-242. [PMID: 39207695 DOI: 10.1007/978-3-031-55529-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neural plasticity can be defined as the ability of neural circuits to be shaped by external and internal factors. It provides the brain with a capacity for functional and morphological remodelling, with many lines of evidence indicating that these changes are vital for learning and memory formation. The basis of this brain plasticity resides in activity- and experience-driven modifications of synaptic strength, including synaptic formation, elimination or weakening, as well as of modulation of neuronal population, which drive the structural reorganization of neural networks. Recent evidence indicates that brain-resident glial cells actively participate in these processes, suggesting that mechanisms underlying plasticity in the brain are multifaceted. Establishing the 'tripartite' synapse, the role of astrocytes in modulating synaptic transmission in response to neuronal activity was recognized first. Further redefinition of the synapse as 'quad-partite' followed to acknowledge the contribution of microglia which were revealed to affect numerous brain functions via dynamic interactions with synapses, acting as 'synaptic sensors' that respond to neuronal activity and neurotransmitter release, as well as crosstalk with astrocytes. Early studies identified microglial ability to dynamically survey their local brain environment and established their integral role in the active interfacing of environmental stimuli (both internal and external), with brain plasticity and remodelling. Following the introduction to neurogenesis, this chapter details the role that microglia play in regulating neurogenesis in adulthood, specifically as it relates to learning and memory, as well as factors involved in modulation of microglia. Further, a microglial perspective is introduced for the context of environmental enrichment impact on neurogenesis, learning and memory across states of stress, ageing, disease and injury.
Collapse
Affiliation(s)
- Eva Šimončičová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | | | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
33
|
Ferrucci L, Cantando I, Cordella F, Di Angelantonio S, Ragozzino D, Bezzi P. Microglia at the Tripartite Synapse during Postnatal Development: Implications for Autism Spectrum Disorders and Schizophrenia. Cells 2023; 12:2827. [PMID: 38132147 PMCID: PMC10742295 DOI: 10.3390/cells12242827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Synapses are the fundamental structures of neural circuits that control brain functions and behavioral and cognitive processes. Synapses undergo formation, maturation, and elimination mainly during postnatal development via a complex interplay with neighboring astrocytes and microglia that, by shaping neural connectivity, may have a crucial role in the strengthening and weakening of synaptic functions, that is, the functional plasticity of synapses. Indeed, an increasing number of studies have unveiled the roles of microglia and astrocytes in synapse formation, maturation, and elimination as well as in regulating synaptic function. Over the past 15 years, the mechanisms underlying the microglia- and astrocytes-dependent regulation of synaptic plasticity have been thoroughly studied, and researchers have reported that the disruption of these glial cells in early postnatal development may underlie the cause of synaptic dysfunction that leads to neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia.
Collapse
Affiliation(s)
- Laura Ferrucci
- Department of Physiology and Pharmacology, University of Rome Sapienza, 00185 Rome, Italy; (L.F.); (F.C.); (S.D.A.); (D.R.)
| | - Iva Cantando
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland;
| | - Federica Cordella
- Department of Physiology and Pharmacology, University of Rome Sapienza, 00185 Rome, Italy; (L.F.); (F.C.); (S.D.A.); (D.R.)
- Center for Life Nano- & Neuro-Science, IIT, 00161 Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, University of Rome Sapienza, 00185 Rome, Italy; (L.F.); (F.C.); (S.D.A.); (D.R.)
- Center for Life Nano- & Neuro-Science, IIT, 00161 Rome, Italy
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, University of Rome Sapienza, 00185 Rome, Italy; (L.F.); (F.C.); (S.D.A.); (D.R.)
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Paola Bezzi
- Department of Physiology and Pharmacology, University of Rome Sapienza, 00185 Rome, Italy; (L.F.); (F.C.); (S.D.A.); (D.R.)
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland;
| |
Collapse
|
34
|
Chen J, Wang T, Zhou Y, Hong Y, Zhang S, Zhou Z, Jiang A, Liu D. Microglia trigger the structural plasticity of GABAergic neurons in the hippocampal CA1 region of a lipopolysaccharide-induced neuroinflammation model. Exp Neurol 2023; 370:114565. [PMID: 37806513 DOI: 10.1016/j.expneurol.2023.114565] [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: 05/04/2023] [Revised: 09/23/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
It is well-established that microglia-mediated neuroinflammatory response involves numerous neuropsychiatric and neurodegenerative diseases. While the role of microglia in excitatory synaptic transmission has been widely investigated, the impact of innate immunity on the structural plasticity of GABAergic inhibitory synapses is not well understood. To investigate this, we established an inflammation model using lipopolysaccharide (LPS) and observed a prolonged microglial response in the hippocampal CA1 region of mice, which was associated with cognitive deficits in the open field test, Y-maze test, and novel object recognition test. Furthermore, we found an increased abundance of GABAergic interneurons and GABAergic synapse formation in the hippocampal CA1 region. The cognitive impairment caused by LPS injection could be reversed by blocking GABA receptor activity with (-)-Bicuculline methiodide. These findings suggest that the upregulation of GABAergic synapses induced by LPS-mediated microglial activation leads to cognitive dysfunction. Additionally, the depletion of microglia by PLX3397 resulted in a decrease in GABAergic interneurons and GABAergic inhibitory synapses, which blocked the cognitive decline induced by LPS. In conclusion, our findings indicate that excessive reinforcement of GABAergic inhibitory synapse formation via microglial activation contributes to LPS-induced cognitive impairment.
Collapse
Affiliation(s)
- Juan Chen
- School of Mental Health, Bengbu Medical College, Bengbu 233030, China
| | - Tao Wang
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Yuting Zhou
- School of Mental Health, Bengbu Medical College, Bengbu 233030, China
| | - Yiming Hong
- School of Mental Health, Bengbu Medical College, Bengbu 233030, China
| | - Shiyong Zhang
- School of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - Zhongtao Zhou
- School of Nursing, Bengbu Medical College, Bengbu 233030, China
| | - Ao Jiang
- School of Mental Health, Bengbu Medical College, Bengbu 233030, China
| | - Danyang Liu
- Department of Ophthalmology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
35
|
Bogue MA, Ball RL, Walton DO, Dunn MH, Kolishovski G, Berger A, Lamoureux A, Grubb SC, Gerring M, Kim M, Liang H, Emerson J, Stearns T, He H, Mukherjee G, Bluis J, Davis S, Desai S, Sundberg B, Kadakkuzha B, Kunde-Ramamoorthy G, Philip VM, Chesler EJ. Mouse phenome database: curated data repository with interactive multi-population and multi-trait analyses. Mamm Genome 2023; 34:509-519. [PMID: 37581698 PMCID: PMC10627943 DOI: 10.1007/s00335-023-10014-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/25/2023] [Indexed: 08/16/2023]
Abstract
The Mouse Phenome Database continues to serve as a curated repository and analysis suite for measured attributes of members of diverse mouse populations. The repository includes annotation to community standard ontologies and guidelines, a database of allelic states for 657 mouse strains, a collection of protocols, and analysis tools for flexible, interactive, user directed analyses that increasingly integrates data across traits and populations. The database has grown from its initial focus on a standard set of inbred strains to include heterogeneous mouse populations such as the Diversity Outbred and mapping crosses and well as Collaborative Cross, Hybrid Mouse Diversity Panel, and recombinant inbred strains. Most recently the system has expanded to include data from the International Mouse Phenotyping Consortium. Collectively these data are accessible by API and provided with an interactive tool suite that enables users' persistent selection, storage, and operation on collections of measures. The tool suite allows basic analyses, advanced functions with dynamic visualization including multi-population meta-analysis, multivariate outlier detection, trait pattern matching, correlation analyses and other functions. The data resources and analysis suite provide users a flexible environment in which to explore the basis of phenotypic variation in health and disease across the lifespan.
Collapse
Affiliation(s)
- Molly A Bogue
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA.
| | - Robyn L Ball
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - David O Walton
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Matthew H Dunn
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | | | | - Anna Lamoureux
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Stephen C Grubb
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Matthew Gerring
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Matthew Kim
- University of British Columbia, Vancouver, BC, Canada
| | - Hongping Liang
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Jake Emerson
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Timothy Stearns
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Hao He
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | | - John Bluis
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Sara Davis
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Sejal Desai
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | - Beth Sundberg
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | | | | - Vivek M Philip
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | |
Collapse
|
36
|
Yan L, Xuan FL, Chen S, Gou M, Chen W, Li Y, Wang Z, Wang L, Xie T, Fan F, Zharkovsky A, Tan Y, Tian L. Replenished microglia partially rescue schizophrenia-related stress response. Front Cell Neurosci 2023; 17:1254923. [PMID: 37771931 PMCID: PMC10522857 DOI: 10.3389/fncel.2023.1254923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/15/2023] [Indexed: 09/30/2023] Open
Abstract
Background Microglia play an important role in the maintenance of brain and behavioral homeostasis. The protective effect of microglial replenishment was reported in neurological diseases, but whether microglial therapy would benefit psychiatric disorders such as schizophrenia has been unclear. As schizophrenia is a stress-vulnerable disorder and psychosocial stress promotes inflammation and microglial activation, we aim to understand how microglial replenishment works in stress-associated schizophrenia. Methods We used a CSF1R-mediated pharmacological approach to study repopulated microglia (repMg) in a cohort of mice (n = 10/group) undergoing chronic unpredictable stress (CUS). We further studied a cohort of first-episode schizophrenia (FES, n = 74) patients who had higher perceived stress scores (PSS) than healthy controls (HCs, n = 68). Results Reborn microglia attenuated CUS-induced learned hopelessness and social withdrawal but not anxiety in mice. Compared to control, CUS- or repMg-induced differentially expressed genes (DEGs) in the prefrontal cortex regulated nervous system development and axonal guidance. CUS also caused microglial hyper-ramification and increased engulfment of synaptophysin and vesicular glutamate transporter-2 by microglia and astrocytes, which were recovered in CUS + repMg (all p < 0.05). Moreover, FES patients had smaller hippocampal fimbria than HCs (p < 1e-7), which were negatively associated with PSS (r = -0.397, p = 0.003). Blood DEGs involved in immune system development were also associated with PSS and the right fimbria more prominently in FES patients than HCs (Zr, p < 0.0001). The KCNQ1 was a partial mediator between PSS and fimbria size (β = -0.442, 95% CI: -1.326 ~ -0.087). Conclusion Microglial replenishment may potentially benefit psychiatric disorders such as schizophrenia.
Collapse
Affiliation(s)
- Ling Yan
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Fang-Ling Xuan
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Song Chen
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Mengzhuang Gou
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Wenjin Chen
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Yanli Li
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Zhiren Wang
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Leilei Wang
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Ting Xie
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Fengmei Fan
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Alexander Zharkovsky
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Yunlong Tan
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Li Tian
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| |
Collapse
|
37
|
Kulczyńska-Przybik A, Dulewicz M, Doroszkiewicz J, Borawska R, Słowik A, Zetterberg H, Hanrieder J, Blennow K, Mroczko B. The Relationships between Cerebrospinal Fluid Glial (CXCL12, CX3CL, YKL-40) and Synaptic Biomarkers (Ng, NPTXR) in Early Alzheimer's Disease. Int J Mol Sci 2023; 24:13166. [PMID: 37685973 PMCID: PMC10487764 DOI: 10.3390/ijms241713166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
In addition to amyloid and tau pathology in the central nervous system (CNS), inflammatory processes and synaptic dysfunction are highly important mechanisms involved in the development and progression of dementia diseases. In the present study, we conducted a comparative analysis of selected pro-inflammatory proteins in the CNS with proteins reflecting synaptic damage and core biomarkers in mild cognitive impairment (MCI) and early Alzheimer's disease (AD). To our knowledge, no studies have yet compared CXCL12 and CX3CL1 with markers of synaptic disturbance in cerebrospinal fluid (CSF) in the early stages of dementia. The quantitative assessment of selected proteins in the CSF of patients with MCI, AD, and non-demented controls (CTRL) was performed using immunoassays (single- and multiplex techniques). In this study, increased CSF concentration of CX3CL1 in MCI and AD patients correlated positively with neurogranin (r = 0.74; p < 0.001, and r = 0.40; p = 0.020, respectively), ptau181 (r = 0.49; p = 0.040), and YKL-40 (r = 0.47; p = 0.050) in MCI subjects. In addition, elevated CSF levels of CXCL12 in the AD group were significantly associated with mini-mental state examination score (r = -0.32; p = 0.040). We found significant evidence to support an association between CX3CL1 and neurogranin, already in the early stages of cognitive decline. Furthermore, our findings indicate that CXCL12 might be a useful marker for tract severity of cognitive impairment.
Collapse
Affiliation(s)
| | - Maciej Dulewicz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Renata Borawska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Agnieszka Słowik
- Department of Neurology, Jagiellonian University, 30-688 Kraków, Poland
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London WC1N 3AR, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792-2460, USA
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- SciLifeLab, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 431 80 Mölndal, Sweden
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
38
|
D'Antoni C, Mautone L, Sanchini C, Tondo L, Grassmann G, Cidonio G, Bezzi P, Cordella F, Di Angelantonio S. Unlocking Neural Function with 3D In Vitro Models: A Technical Review of Self-Assembled, Guided, and Bioprinted Brain Organoids and Their Applications in the Study of Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2023; 24:10762. [PMID: 37445940 DOI: 10.3390/ijms241310762] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Understanding the complexities of the human brain and its associated disorders poses a significant challenge in neuroscience. Traditional research methods have limitations in replicating its intricacies, necessitating the development of in vitro models that can simulate its structure and function. Three-dimensional in vitro models, including organoids, cerebral organoids, bioprinted brain models, and functionalized brain organoids, offer promising platforms for studying human brain development, physiology, and disease. These models accurately replicate key aspects of human brain anatomy, gene expression, and cellular behavior, enabling drug discovery and toxicology studies while providing insights into human-specific phenomena not easily studied in animal models. The use of human-induced pluripotent stem cells has revolutionized the generation of 3D brain structures, with various techniques developed to generate specific brain regions. These advancements facilitate the study of brain structure development and function, overcoming previous limitations due to the scarcity of human brain samples. This technical review provides an overview of current 3D in vitro models of the human cortex, their development, characterization, and limitations, and explores the state of the art and future directions in the field, with a specific focus on their applications in studying neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Chiara D'Antoni
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Lorenza Mautone
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Caterina Sanchini
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Lucrezia Tondo
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Greta Grassmann
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, 00185 Rome, Italy
| | - Gianluca Cidonio
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Paola Bezzi
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Department of Fundamental Neurosciences, University of Lausanne, 1011 Lausanne, Switzerland
| | - Federica Cordella
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- and Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- D-Tails s.r.l., 00165 Rome, Italy
| |
Collapse
|
39
|
Sun Y, Che J, Zhang J. Emerging non-proinflammatory roles of microglia in healthy and diseased brains. Brain Res Bull 2023; 199:110664. [PMID: 37192719 DOI: 10.1016/j.brainresbull.2023.110664] [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: 07/14/2022] [Revised: 04/04/2023] [Accepted: 05/13/2023] [Indexed: 05/18/2023]
Abstract
Microglia, the resident myeloid cells of the central nervous system, are the first line of defense against foreign pathogens, thereby confining the extent of brain injury. However, the role of microglia is not limited to macrophage-like functions. In addition to proinflammatory response mediation, microglia are involved in neurodevelopmental remodeling and homeostatic maintenance in the absence of disease. An increasing number of studies have also elucidated microglia-mediated regulation of tumor growth and neural repair in diseased brains. Here, we review the non-proinflammatory roles of microglia, with the aim of promoting a deeper understanding of the functions of microglia in healthy and diseased brains and contributing to the development of novel therapeutics that target microglia in neurological disorders.
Collapse
Affiliation(s)
- Yinying Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, 200032, Shanghai China.
| | - Ji Che
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, 200032, Shanghai China.
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, 200032, Shanghai China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai China.
| |
Collapse
|
40
|
Pan Q, Yan P, Kim AB, Xiao Q, Pandey G, Haecker H, Epelman S, Diwan A, Lee JM, DeSelm CJ. Chimeric Antigen Receptor Macrophages Target and Resorb Amyloid Plaques in a Mouse Model of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538637. [PMID: 37162824 PMCID: PMC10168376 DOI: 10.1101/2023.04.28.538637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Substantial evidence suggests a role for immunotherapy in treating Alzheimer's disease (AD). Several monoclonal antibodies targeting aggregated forms of beta amyloid (Aβ), have been shown to reduce amyloid plaques and in some cases, mitigate cognitive decline in early-stage AD patients. We sought to determine if genetically engineered macrophages could improve the targeting and degradation of amyloid plaques. Chimeric antigen receptor macrophages (CAR-Ms), which show promise as a cancer treatment, are an appealing strategy to enhance target recognition and phagocytosis of amyloid plaques in AD. We genetically engineered macrophages to express a CAR containing the anti-amyloid antibody aducanumab as the external domain and the Fc receptor signaling domain internally. CAR-Ms recognize and degrade Aβ in vitro and on APP/PS1 brain slices ex vivo; however, when injected intrahippocampally, these first-generation CAR-Ms have limited persistence and fail to reduce plaque load. We overcame this limitation by creating CAR-Ms that secrete M-CSF and self-maintain without exogenous cytokines. These CAR-Ms have greater survival in the brain niche, and significantly reduce plaque load locally in vivo. These proof-of-principle studies demonstrate that CAR-Ms, previously only applied to cancer, may be utilized to target and degrade unwanted materials, such as amyloid plaques in the brains of AD mice.
Collapse
Affiliation(s)
- Qiuyun Pan
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ping Yan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexander B. Kim
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Bursky Center for Human Immunology and Immunotherapy, Washington University School of Medicine, St. Louis, MO, USA
| | - Qingli Xiao
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Gaurav Pandey
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Bursky Center for Human Immunology and Immunotherapy, Washington University School of Medicine, St. Louis, MO, USA
| | - Hans Haecker
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Slava Epelman
- Department of Medicine, Division of Cardiology, Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Abhinav Diwan
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
- Medicine Service, Saint Louis VA Medical Center, St. Louis, MO, USA
- Departments of Medicine, Cell Biology and Physiology, Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Carl J. DeSelm
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Bursky Center for Human Immunology and Immunotherapy, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
41
|
VanderZwaag J, Halvorson T, Dolhan K, Šimončičová E, Ben-Azu B, Tremblay MÈ. The Missing Piece? A Case for Microglia's Prominent Role in the Therapeutic Action of Anesthetics, Ketamine, and Psychedelics. Neurochem Res 2023; 48:1129-1166. [PMID: 36327017 DOI: 10.1007/s11064-022-03772-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
There is much excitement surrounding recent research of promising, mechanistically novel psychotherapeutics - psychedelic, anesthetic, and dissociative agents - as they have demonstrated surprising efficacy in treating central nervous system (CNS) disorders, such as mood disorders and addiction. However, the mechanisms by which these drugs provide such profound psychological benefits are still to be fully elucidated. Microglia, the CNS's resident innate immune cells, are emerging as a cellular target for psychiatric disorders because of their critical role in regulating neuroplasticity and the inflammatory environment of the brain. The following paper is a review of recent literature surrounding these neuropharmacological therapies and their demonstrated or hypothesized interactions with microglia. Through investigating the mechanism of action of psychedelics, such as psilocybin and lysergic acid diethylamide, ketamine, and propofol, we demonstrate a largely under-investigated role for microglia in much of the emerging research surrounding these pharmacological agents. Among others, we detail sigma-1 receptors, serotonergic and γ-aminobutyric acid signalling, and tryptophan metabolism as pathways through which these agents modulate microglial phagocytic activity and inflammatory mediator release, inducing their therapeutic effects. The current review includes a discussion on future directions in the field of microglial pharmacology and covers bidirectional implications of microglia and these novel pharmacological agents in aging and age-related disease, glial cell heterogeneity, and state-of-the-art methodologies in microglial research.
Collapse
Affiliation(s)
- Jared VanderZwaag
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Torin Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Vancouver, BC, Canada
- Department of Biology, University of Victoria, Vancouver, BC, Canada
| | - Eva Šimončičová
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de médecine moléculaire, Université Laval, Québec City, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
42
|
Manickam V, Gawande DY, Stothert AR, Clayman AC, Batalkina L, Warchol ME, Ohlemiller KK, Kaur T. Macrophages Promote Repair of Inner Hair Cell Ribbon Synapses following Noise-Induced Cochlear Synaptopathy. J Neurosci 2023; 43:2075-2089. [PMID: 36810227 PMCID: PMC10039750 DOI: 10.1523/jneurosci.1273-22.2023] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023] Open
Abstract
Resident cochlear macrophages rapidly migrate into the inner hair cell synaptic region and directly contact the damaged synaptic connections after noise-induced synaptopathy. Eventually, such damaged synapses are spontaneously repaired, but the precise role of macrophages in synaptic degeneration and repair remains unknown. To address this, cochlear macrophages were eliminated using colony stimulating factor 1 receptor (CSF1R) inhibitor, PLX5622. Sustained treatment with PLX5622 in CX3CR1 GFP/+ mice of both sexes led to robust elimination of resident macrophages (∼94%) without significant adverse effects on peripheral leukocytes, cochlear function, and structure. At 1 day (d) post noise exposure of 93 or 90 dB SPL for 2 hours, the degree of hearing loss and synapse loss were comparable in the presence and absence of macrophages. At 30 d after exposure, damaged synapses appeared repaired in the presence of macrophages. However, in the absence of macrophages, such synaptic repair was significantly reduced. Remarkably, on cessation of PLX5622 treatment, macrophages repopulated the cochlea, leading to enhanced synaptic repair. Elevated auditory brainstem response thresholds and reduced auditory brainstem response Peak 1 amplitudes showed limited recovery in the absence of macrophages but recovered similarly with resident and repopulated macrophages. Cochlear neuron loss was augmented in the absence of macrophages but showed preservation with resident and repopulated macrophages after noise exposure. While the central auditory effects of PLX5622 treatment and microglia depletion remain to be investigated, these data demonstrate that macrophages do not affect synaptic degeneration but are necessary and sufficient to restore cochlear synapses and function after noise-induced synaptopathy.SIGNIFICANCE STATEMENT The synaptic connections between cochlear inner hair cells and spiral ganglion neurons can be lost because of noise over exposure or biological aging. This loss may represent the most common causes of sensorineural hearing loss also known as hidden hearing loss. Synaptic loss results in degradation of auditory information, leading to difficulty in listening in noisy environments and other auditory perceptual disorders. We demonstrate that resident macrophages of the cochlea are necessary and sufficient to restore synapses and function following synaptopathic noise exposure. Our work reveals a novel role for innate-immune cells, such as macrophages in synaptic repair, that could be harnessed to regenerate lost ribbon synapses in noise- or age-linked cochlear synaptopathy, hidden hearing loss, and associated perceptual anomalies.
Collapse
Affiliation(s)
- Vijayprakash Manickam
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, Nebraska 68178
| | - Dinesh Y Gawande
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, Nebraska 68178
| | - Andrew R Stothert
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, Nebraska 68178
| | - Anna C Clayman
- Department of Otolaryngology, School of Medicine, Washington University, St. Louis, Missouri 63110
| | - Lyudmila Batalkina
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, Nebraska 68178
| | - Mark E Warchol
- Department of Otolaryngology, School of Medicine, Washington University, St. Louis, Missouri 63110
| | - Kevin K Ohlemiller
- Department of Otolaryngology, School of Medicine, Washington University, St. Louis, Missouri 63110
| | - Tejbeer Kaur
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, Nebraska 68178
| |
Collapse
|
43
|
Rosito M, Sanchini C, Gosti G, Moreno M, De Panfilis S, Giubettini M, Debellis D, Catalano F, Peruzzi G, Marotta R, Indrieri A, De Leonibus E, De Stefano ME, Ragozzino D, Ruocco G, Di Angelantonio S, Bartolini F. Microglia reactivity entails microtubule remodeling from acentrosomal to centrosomal arrays. Cell Rep 2023; 42:112104. [PMID: 36787220 PMCID: PMC10423306 DOI: 10.1016/j.celrep.2023.112104] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 12/02/2022] [Accepted: 01/27/2023] [Indexed: 02/15/2023] Open
Abstract
Microglia reactivity entails a large-scale remodeling of cellular geometry, but the behavior of the microtubule cytoskeleton during these changes remains unexplored. Here we show that activated microglia provide an example of microtubule reorganization from a non-centrosomal array of parallel and stable microtubules to a radial array of more dynamic microtubules. While in the homeostatic state, microglia nucleate microtubules at Golgi outposts, and activating signaling induces recruitment of nucleating material nearby the centrosome, a process inhibited by microtubule stabilization. Our results demonstrate that a hallmark of microglia reactivity is a striking remodeling of the microtubule cytoskeleton and suggest that while pericentrosomal microtubule nucleation may serve as a distinct marker of microglia activation, inhibition of microtubule dynamics may provide a different strategy to reduce microglia reactivity in inflammatory disease.
Collapse
Affiliation(s)
- Maria Rosito
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy; Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy
| | - Caterina Sanchini
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy; Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy
| | - Giorgio Gosti
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy; Soft and Living Matter Laboratory, Institute of Nanotechnology, Consiglio Nazionale delle Ricerche, 00185 Rome, Italy
| | - Manuela Moreno
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy
| | - Simone De Panfilis
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy
| | | | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Federico Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Giovanna Peruzzi
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy
| | - Roberto Marotta
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Alessia Indrieri
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy; Institute for Genetic and Biomedical Research, National Research Council, 20090 Milan, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy; Institute of Biochemistry and Cellular Biology, National Research Council, 00015 Rome, Italy
| | - Maria Egle De Stefano
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185 Rome, Italy
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy; Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00179 Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy; Department of Physics, Sapienza University, 00185 Rome, Italy
| | - Silvia Di Angelantonio
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, 00161 Rome, Italy; Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy; D-Tails s.r.l, 00165 Rome, Italy.
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
44
|
Pereira CPM, Francis-Oliveira J, Singulani MP, Ferreira AFF, Britto LRG. Microglial depletion exacerbates motor impairment and dopaminergic neuron loss in a 6-OHDA model of Parkinson's disease. J Neuroimmunol 2023; 375:578019. [PMID: 36681049 DOI: 10.1016/j.jneuroim.2023.578019] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/28/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
6-hydroxydopamine (6-OHDA) is a common neurotoxin used to induce Parkinson's disease (PD) in mice, exerting neurotoxic effects through the production of reactive oxygen species and microglial activation. However, the role of microglia in PD is still not clear, with contradictory reports showing neuroprotection or exacerbation of neuronal death. Microglial depletion aggravates motor coordination impairments and reduces tyrosine hydroxylase positive neurons in the substantia nigra pars compacta. Moreover, MeCP2 and Adora1 genes expression were downregulated, suggesting they may be involved in the neurodegenerative process. This study highlights that microglia plays a protective role in dopaminergic neuron survival during the initial phase of PD, and the investigation of the mechanisms of this effect in future studies will help elucidate the pathophysiology of PD.
Collapse
Affiliation(s)
- Carolina Parga Martins Pereira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil; Department of Neurobiology and Behavior, Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, USA.
| | - José Francis-Oliveira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil; Departament of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, USA
| | - Monique Patricio Singulani
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Flávia Fernandes Ferreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Roberto G Britto
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
Henning L, Antony H, Breuer A, Müller J, Seifert G, Audinat E, Singh P, Brosseron F, Heneka MT, Steinhäuser C, Bedner P. Reactive microglia are the major source of tumor necrosis factor alpha and contribute to astrocyte dysfunction and acute seizures in experimental temporal lobe epilepsy. Glia 2023; 71:168-186. [PMID: 36373840 DOI: 10.1002/glia.24265] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/09/2022]
Abstract
Extensive microglia reactivity has been well described in human and experimental temporal lobe epilepsy (TLE). To date, however, it is not clear whether and based on which molecular mechanisms microglia contribute to the development and progression of focal epilepsy. Astroglial gap junction coupled networks play an important role in regulating neuronal activity and loss of interastrocytic coupling causally contributes to TLE. Here, we show in the unilateral intracortical kainate (KA) mouse model of TLE that reactive microglia are primary producers of tumor necrosis factor (TNF)α and contribute to astrocyte dysfunction and severity of status epilepticus (SE). Immunohistochemical analyses revealed pronounced and persistent microglia reactivity, which already started 4 h after KA-induced SE. Partial depletion of microglia using a colony stimulating factor 1 receptor inhibitor prevented early astrocyte uncoupling and attenuated the severity of SE, but increased the mortality of epileptic mice following surgery. Using microglia-specific inducible TNFα knockout mice we identified microglia as the major source of TNFα during early epileptogenesis. Importantly, microglia-specific TNFα knockout prevented SE-induced gap junction uncoupling in astrocytes. Continuous telemetric EEG recordings revealed that during the first 4 weeks after SE induction, microglial TNFα did not significantly contribute to spontaneous generalized seizure activity. Moreover, the absence of microglial TNFα did not affect the development of hippocampal sclerosis but attenuated gliosis. Taken together, these data implicate reactive microglia in astrocyte dysfunction and network hyperexcitability after an epileptogenic insult.
Collapse
Affiliation(s)
- Lukas Henning
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Henrike Antony
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Annika Breuer
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Julia Müller
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Gerald Seifert
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Etienne Audinat
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | | | | | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
46
|
Maurya SK, Gupta S, Mishra R. Transcriptional and epigenetic regulation of microglia in maintenance of brain homeostasis and neurodegeneration. Front Mol Neurosci 2023; 15:1072046. [PMID: 36698776 PMCID: PMC9870594 DOI: 10.3389/fnmol.2022.1072046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023] Open
Abstract
The emerging role of microglia in brain homeostasis, neurodegeneration, and neurodevelopmental disorders has attracted considerable interest. In addition, recent developments in microglial functions and associated pathways have shed new light on their fundamental role in the immunological surveillance of the brain. Understanding the interconnections between microglia, neurons, and non-neuronal cells have opened up additional avenues for research in this evolving field. Furthermore, the study of microglia at the transcriptional and epigenetic levels has enhanced our knowledge of these native brain immune cells. Moreover, exploring various facets of microglia biology will facilitate the early detection, treatment, and management of neurological disorders. Consequently, the present review aimed to provide comprehensive insight on microglia biology and its influence on brain development, homeostasis, management of disease, and highlights microglia as potential therapeutic targets in neurodegenerative and neurodevelopmental diseases.
Collapse
Affiliation(s)
- Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India,*Correspondence: Shashank Kumar Maurya, ;
| | - Suchi Gupta
- Tech Cell Innovations Private Limited, Centre for Medical Innovation and Entrepreneurship (CMIE), All India Institute of Medical Sciences, New Delhi, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
47
|
Fractalkine/CX3CR1-Dependent Modulation of Synaptic and Network Plasticity in Health and Disease. Neural Plast 2023; 2023:4637073. [PMID: 36644710 PMCID: PMC9833910 DOI: 10.1155/2023/4637073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 01/06/2023] Open
Abstract
CX3CR1 is a G protein-coupled receptor that is expressed exclusively by microglia within the brain parenchyma. The only known physiological CX3CR1 ligand is the chemokine fractalkine (FKN), which is constitutively expressed in neuronal cell membranes and tonically released by them. Through its key role in microglia-neuron communication, the FKN/CX3CR1 axis regulates microglial state, neuronal survival, synaptic plasticity, and a variety of synaptic functions, as well as neuronal excitability via cytokine release modulation, chemotaxis, and phagocytosis. Thus, the absence of CX3CR1 or any failure in the FKN/CX3CR1 axis has been linked to alterations in different brain functions, including changes in synaptic and network plasticity in structures such as the hippocampus, cortex, brainstem, and spinal cord. Since synaptic plasticity is a basic phenomenon in neural circuit integration and adjustment, here, we will review its modulation by the FKN/CX3CR1 axis in diverse brain circuits and its impact on brain function and adaptation in health and disease.
Collapse
|
48
|
Picard K, Corsi G, Decoeur F, Di Castro MA, Bordeleau M, Persillet M, Layé S, Limatola C, Tremblay MÈ, Nadjar A. Microglial homeostasis disruption modulates non-rapid eye movement sleep duration and neuronal activity in adult female mice. Brain Behav Immun 2023; 107:153-164. [PMID: 36202169 DOI: 10.1016/j.bbi.2022.09.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 09/12/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022] Open
Abstract
Sleep is a natural physiological state, tightly regulated through several neuroanatomical and neurochemical systems, which is essential to maintain physical and mental health. Recent studies revealed that the functions of microglia, the resident immune cells of the brain, differ along the sleep-wake cycle. Inflammatory cytokines, such as interleukin-1β and tumor necrosis factor-α, mainly produced by microglia in the brain, are also well-known to promote sleep. However, the contributing role of microglia on sleep regulation remains largely elusive, even more so in females. Given the higher prevalence of various sleep disorders in women, we aimed to determine the role of microglia in regulating the sleep-wake cycle specifically in female mice. Microglia were depleted in adult female mice with inhibitors of the colony-stimulating factor 1 receptor (CSF1R) (PLX3397 or PLX5622), which is required for microglial population maintenance. This led to a 65-73% reduction of the microglial population, as confirmed by immunofluorescence staining against IBA1 (marker of microglia/macrophages) and TMEM119 (microglia-specific marker) in the reticular nucleus of the thalamus and primary motor cortex. The spontaneous sleep-wake cycle was evaluated at steady-state, during microglial homeostasis disruption and after complete microglial repopulation, upon cessation of treatment with the inhibitors of CSF1R, using electroencephalography (EEG) and electromyography (EMG). We found that microglia-depleted female mice spent more time in non-rapid eye movement (NREM) sleep and had an increased number of NREM sleep episodes, which was partially restored after microglial total repopulation. To determine whether microglia could regulate sleep locally by modulating synaptic transmission, we used patch clamp to record spontaneous activity of pyramidal neurons in the primary motor cortex, which showed an increase of excitatory synaptic transmission during the dark phase. These changes in neuronal activity were modulated by microglial depletion in a phase-dependent manner. Altogether, our results indicate that microglia are involved in the sleep regulation of female mice, further strengthening their potential implication in the development and/or progression of sleep disorders. Furthermore, our findings indicate that microglial repopulation can contribute to normalizing sleep alterations caused by their partial depletion.
Collapse
Affiliation(s)
- Katherine Picard
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Giorgio Corsi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Fanny Decoeur
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | | | - Maude Bordeleau
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Marine Persillet
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Department of Neurophysiology, Neuropharmacology, Inflammaging, IRCCS Neuromed, Pozzilli, Italy
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Agnès Nadjar
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; Institut Universitaire de France (IUF), France.
| |
Collapse
|
49
|
Machado da Silva MC, Iglesias LP, Candelario-Jalil E, Khoshbouei H, Moreira FA, de Oliveira ACP. Role of Microglia in Psychostimulant Addiction. Curr Neuropharmacol 2023; 21:235-259. [PMID: 36503452 PMCID: PMC10190137 DOI: 10.2174/1570159x21666221208142151] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022] Open
Abstract
The use of psychostimulant drugs can modify brain function by inducing changes in the reward system, mainly due to alterations in dopaminergic and glutamatergic transmissions in the mesocorticolimbic pathway. However, the etiopathogenesis of addiction is a much more complex process. Previous data have suggested that microglia and other immune cells are involved in events associated with neuroplasticity and memory, which are phenomena that also occur in addiction. Nevertheless, how dependent is the development of addiction on the activity of these cells? Although the mechanisms are not known, some pathways may be involved. Recent data have shown psychoactive substances may act directly on immune cells, alter their functions and induce various inflammatory mediators that modulate synaptic activity. These could, in turn, be involved in the pathological alterations that occur in substance use disorder. Here, we extensively review the studies demonstrating how cocaine and amphetamines modulate microglial number, morphology, and function. We also describe the effect of these substances in the production of inflammatory mediators and a possible involvement of some molecular signaling pathways, such as the toll-like receptor 4. Although the literature in this field is scarce, this review compiles the knowledge on the neuroimmune axis that is involved in the pathogenesis of addiction, and suggests some pharmacological targets for the development of pharmacotherapy.
Collapse
Affiliation(s)
- Maria Carolina Machado da Silva
- Department of Pharmacology, Neuropharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil;
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lia Parada Iglesias
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fabrício Araujo Moreira
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
50
|
De Felice E, Gonçalves de Andrade E, Golia MT, González Ibáñez F, Khakpour M, Di Castro MA, Garofalo S, Di Pietro E, Benatti C, Brunello N, Tascedda F, Kaminska B, Limatola C, Ragozzino D, Tremblay ME, Alboni S, Maggi L. Microglial diversity along the hippocampal longitudinal axis impacts synaptic plasticity in adult male mice under homeostatic conditions. J Neuroinflammation 2022; 19:292. [PMID: 36482444 PMCID: PMC9730634 DOI: 10.1186/s12974-022-02655-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is a plastic brain area that shows functional segregation along its longitudinal axis, reflected by a higher level of long-term potentiation (LTP) in the CA1 region of the dorsal hippocampus (DH) compared to the ventral hippocampus (VH), but the mechanisms underlying this difference remain elusive. Numerous studies have highlighted the importance of microglia-neuronal communication in modulating synaptic transmission and hippocampal plasticity, although its role in physiological contexts is still largely unknown. We characterized in depth the features of microglia in the two hippocampal poles and investigated their contribution to CA1 plasticity under physiological conditions. We unveiled the influence of microglia in differentially modulating the amplitude of LTP in the DH and VH, showing that minocycline or PLX5622 treatment reduced LTP amplitude in the DH, while increasing it in the VH. This was recapitulated in Cx3cr1 knockout mice, indicating that microglia have a key role in setting the conditions for plasticity processes in a region-specific manner, and that the CX3CL1-CX3CR1 pathway is a key element in determining the basal level of CA1 LTP in the two regions. The observed LTP differences at the two poles were associated with transcriptional changes in the expression of genes encoding for Il-1, Tnf-α, Il-6, and Bdnf, essential players of neuronal plasticity. Furthermore, microglia in the CA1 SR region showed an increase in soma and a more extensive arborization, an increased prevalence of immature lysosomes accompanied by an elevation in mRNA expression of phagocytic markers Mertk and Cd68 and a surge in the expression of microglial outward K+ currents in the VH compared to DH, suggesting a distinct basal phenotypic state of microglia across the two hippocampal poles. Overall, we characterized the molecular, morphological, ultrastructural, and functional profile of microglia at the two poles, suggesting that modifications in hippocampal subregions related to different microglial statuses can contribute to dissect the phenotypical aspects of many diseases in which microglia are known to be involved.
Collapse
Affiliation(s)
- E. De Felice
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Gonçalves de Andrade
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. T. Golia
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - F. González Ibáñez
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - M. Khakpour
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. A. Di Castro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - S. Garofalo
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Di Pietro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - C. Benatti
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - N. Brunello
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - F. Tascedda
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - B. Kaminska
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - C. Limatola
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur, Sapienza University, Rome, Italy
| | - D. Ragozzino
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy ,grid.417778.a0000 0001 0692 3437Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - M. E. Tremblay
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - S. Alboni
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - L. Maggi
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| |
Collapse
|