1
|
Barbaresi P, Fabri M, Lorenzi T, Sagrati A, Morroni M. Intrinsic organization of the corpus callosum. Front Physiol 2024; 15:1393000. [PMID: 39035452 PMCID: PMC11259024 DOI: 10.3389/fphys.2024.1393000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/16/2024] [Indexed: 07/23/2024] Open
Abstract
The corpus callosum-the largest commissural fiber system connecting the two cerebral hemispheres-is considered essential for bilateral sensory integration and higher cognitive functions. Most studies exploring the corpus callosum have examined either the anatomical, physiological, and neurochemical organization of callosal projections or the functional and/or behavioral aspects of the callosal connections after complete/partial callosotomy or callosal lesion. There are no works that address the intrinsic organization of the corpus callosum. We review the existing information on the activities that take place in the commissure in three sections: I) the topographical and neurochemical organization of the intracallosal fibers, II) the role of glia in the corpus callosum, and III) the role of the intracallosal neurons.
Collapse
Affiliation(s)
- Paolo Barbaresi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Marche Polytechnic University, Ancona, Italy
| | - Mara Fabri
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Teresa Lorenzi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Marche Polytechnic University, Ancona, Italy
| | - Andrea Sagrati
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Manrico Morroni
- Electron Microscopy Unit, Azienda Ospedaliero-Universitaria, Ancona, Italy
| |
Collapse
|
2
|
Yang L, Zhang Y, Yu X, Li D, Liu N, Xue X, Fu J. Periventricular Microglia Polarization and Morphological Changes Accompany NLRP3 Inflammasome-Mediated Neuroinflammation after Hypoxic-Ischemic White Matter Damage in Premature Rats. J Immunol Res 2023; 2023:5149306. [PMID: 37636861 PMCID: PMC10460280 DOI: 10.1155/2023/5149306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/08/2023] [Accepted: 07/13/2023] [Indexed: 08/29/2023] Open
Abstract
White matter damage (WMD) is a primary cause of cerebral palsy and cognitive impairment in preterm infants, and no effective treatments are available. Microglia are a major component of the innate immune system. When activated, they form typical pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes and regulate myelin development and synapse formation. Therefore, they may play a pivotal role in hypoxic-ischemic (HI) WMD. Herein, we investigated neural inflammation and long-term microglia phenotypic polarization in a neonatal rat model of hypoxia-ischemia-induced WMD and elucidated the underlying pathophysiological processes. We exposed 3-day-old (P3) Sprague-Dawley rats to hypoxia (8% oxygen) for 2.5 hr after unilateral common carotid artery ligation. The activation of NLRP3 inflammatory bodies, microglia M1/M2 polarization, myelination, and synaptic development in our model were monitored 7, 14, and 21 days after birth. In addition, the Morris water maze test was performed on postnatal Day 28. We confirmed myelination disturbance in the periventricular white matter, abnormal synaptic development, and behavioral changes in the periventricular area during the development of HI WMD. In addition, we found an association between the occurrence and development of HI WMD and activation of the NLRP3 inflammasome, microglial M1/M2 polarization, and the release of inflammatory factors. NLRP3 inhibition can play an anti-inflammatory role by inhibiting the differentiation of microglia into the M1 phenotype, thereby improving myelination and synapse formation. In conclusion, microglia are key mediators of the inflammatory response and exhibit continuous phenotypic polarization 7-21 days after HI-induced WMD. This finding can potentially lead to a new treatment regimen targeting the phenotypic polarization of microglia early after HI-induced brain injury.
Collapse
Affiliation(s)
- Liu Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
- Department of Pediatrics, The Second Hospital of Dalian Medical University, Dalian 116021, Liaoning, China
| | - Yajun Zhang
- Department of Anesthesiology, Dalian Municipal Maternal and Child Health Care Hospital, Dalian 116021, Liaoning, China
| | - Xuefei Yu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Danni Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Na Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Xindong Xue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| |
Collapse
|
3
|
Carbon Monoxide Modulation of Microglia-Neuron Communication: Anti-Neuroinflammatory and Neurotrophic Role. Mol Neurobiol 2021; 59:872-889. [PMID: 34796462 DOI: 10.1007/s12035-021-02643-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/09/2021] [Indexed: 10/19/2022]
Abstract
Microglia, the 'resident immunocompetent cells' of the central nervous system (CNS), are key players in innate immunity, synaptic refinement and homeostasis. Dysfunctional microglia contribute heavily to creating a toxic inflammatory milieu, a driving factor in the pathophysiology of several CNS disorders. Therefore, strategies to modulate the microglial function are required to tackle exacerbated tissue inflammation. Carbon monoxide (CO), an endogenous gaseous molecule produced by the degradation of haem, has anti-inflammatory, anti-apoptotic, and pro-homeostatic and cytoprotective roles, among others. ALF-826A, a novel molybdenum-based CO-releasing molecule, was used for the assessment of neuron-microglia remote communication. Primary cultures of rat microglia and neurons, or the BV-2 microglial and CAD neuronal murine cell lines, were used to study the microglia-neuron interaction. An approach based on microglial-derived conditioned media in neuronal culture was applied. Medium derived from CO-treated microglia provided indirect neuroprotection against inflammation by limiting the lipopolysaccharide (LPS)-induced expression of reactivity markers (CD11b), the production of reactive oxygen species (ROS) and the secretion of inflammatory factors (TNF-α, nitrites). This consequently prevented neuronal cell death and maintained neuronal morphology. In contrast, in the absence of inflammatory stimulus, conditioned media from CO-treated microglia improved neuronal morphological complexity, which is an indirect manner of assessing neuronal function. Likewise, the microglial medium also prevented neuronal cell death induced by pro-oxidant tert-Butyl hydroperoxide (t-BHP). ALF-826 treatment reinforced microglia secretion of Interleukin-10 (IL-10) and adenosine, mediators that may protect against t-BHP stress in this remote communication model. Chemical inhibition of the adenosine receptors A2A and A1 reverted the CO-derived neuroprotective effect, further highlighting a role for CO in regulating neuron-microglia communication via purinergic signalling. Our findings indicate that CO has a modulatory role on microglia-to-neuron communication, promoting neuroprotection in a non-cell autonomous manner. CO enhances the microglial release of neurotrophic factors and blocks exacerbated microglial inflammation. CO improvement of microglial neurotrophism under non-inflammatory conditions is here described for the first time.
Collapse
|
4
|
Soares NL, Vieira HLA. Microglia at the Centre of Brain Research: Accomplishments and Challenges for the Future. Neurochem Res 2021; 47:218-233. [PMID: 34586585 DOI: 10.1007/s11064-021-03456-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
Microglia are the immune guardians of the central nervous system (CNS), with critical functions in development, maintenance of homeostatic tissue balance, injury and repair. For a long time considered a forgotten 'third element' with basic phagocytic functions, a recent surge in interest, accompanied by technological progress, has demonstrated that these distinct myeloid cells have a wide-ranging importance for brain function. This review reports microglial origins, development, and function in the healthy brain. Moreover, it also targets microglia dysfunction and how it contributes to the progression of several neurological disorders, focusing on particular molecular mechanisms and whether these may present themselves as opportunities for novel, microglia-targeted therapeutic approaches, an ever-enticing prospect. Finally, as it has been recently celebrated 100 years of microglia research, the review highlights key landmarks from the past century and looked into the future. Many challenging problems have arisen, thus it points out some of the most pressing questions and experimental challenges for the ensuing century.
Collapse
Affiliation(s)
- Nuno L Soares
- Chronic Diseases Research Center (CEDOC) - Faculdade de Ciências Médicas/NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria 130, 1169-056, Lisboa, Portugal.
| | - Helena L A Vieira
- Chronic Diseases Research Center (CEDOC) - Faculdade de Ciências Médicas/NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria 130, 1169-056, Lisboa, Portugal.,Department of Chemistry, UCIBIO, Applied Molecular Biosciences Unit, NOVA School of Science and Technology, Universidade Nova de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Lisboa, Portugal
| |
Collapse
|
5
|
Abstract
Microglia are the resident immune cells of the central nervous system. Microglial progenitors are generated in the yolk sac during the early embryonic stage. Once microglia enter the brain primordium, these cells colonize the structure through migration and proliferation during brain development. Microglia account for a minor population among the total cells that constitute the developing cortex, but they can associate with many surrounding neural lineage cells by extending their filopodia and through their broad migration capacity. Of note, microglia change their distribution in a stage-dependent manner in the developing brain: microglia are homogenously distributed in the pallium in the early and late embryonic stages, whereas these cells are transiently absent from the cortical plate (CP) from embryonic day (E) 15 to E16 and colonize the ventricular zone (VZ), subventricular zone (SVZ), and intermediate zone (IZ). Previous studies have reported that microglia positioned in the VZ/SVZ/IZ play multiple roles in neural lineage cells, such as regulating neurogenesis, cell survival and neuronal circuit formation. In addition to microglial functions in the zones in which microglia are replenished, these cells indirectly contribute to the proper maturation of post-migratory neurons by exiting the CP during the mid-embryonic stage. Overall, microglial time-dependent distributional changes are necessary to provide particular functions that are required in specific regions. This review summarizes recent advances in the understanding of microglial colonization and multifaceted functions in the developing brain, especially focusing on the embryonic stage, and discuss the molecular mechanisms underlying microglial behaviors.
Collapse
|
6
|
Sanguino‐Gómez J, Buurstede JC, Abiega O, Fitzsimons CP, Lucassen PJ, Eggen BJL, Lesuis SL, Meijer OC, Krugers HJ. An emerging role for microglia in stress‐effects on memory. Eur J Neurosci 2021; 55:2491-2518. [PMID: 33724565 PMCID: PMC9373920 DOI: 10.1111/ejn.15188] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/13/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
Stressful experiences evoke, among others, a rapid increase in brain (nor)epinephrine (NE) levels and a slower increase in glucocorticoid hormones (GCs) in the brain. Microglia are key regulators of neuronal function and contain receptors for NE and GCs. These brain cells may therefore potentially be involved in modulating stress effects on neuronal function and learning and memory. In this review, we discuss that stress induces (1) an increase in microglial numbers as well as (2) a shift toward a pro‐inflammatory profile. These microglia have (3) impaired crosstalk with neurons and (4) disrupted glutamate signaling. Moreover, microglial immune responses after stress (5) alter the kynurenine pathway through metabolites that impair glutamatergic transmission. All these effects could be involved in the impairments in memory and in synaptic plasticity caused by (prolonged) stress, implicating microglia as a potential novel target in stress‐related memory impairments.
Collapse
Affiliation(s)
| | - Jacobus C. Buurstede
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Oihane Abiega
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Carlos P. Fitzsimons
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| | - Bart J. L. Eggen
- Department of Biomedical Sciences of Cells & Systems Section Molecular Neurobiology University of Groningen University Medical Center Groningen Groningen The Netherlands
| | - Sylvie L. Lesuis
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
- Program in Neurosciences and Mental Health Hospital for Sick Children Toronto ON Canada
| | - Onno C. Meijer
- Department of Medicine Division of Endocrinology Leiden University Medical Center Leiden The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group SILS‐CNS University of Amsterdam Amsterdam The Netherlands
| |
Collapse
|
7
|
Neuroprotective function of microglia in the developing brain. Neuronal Signal 2021; 5:NS20200024. [PMID: 33532089 PMCID: PMC7823182 DOI: 10.1042/ns20200024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 12/16/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system and are important for immune processes. Besides their classical roles in pathological conditions, these cells also dynamically interact with neurons and influence their structure and function in physiological conditions. Recent evidence revealed their role in healthy brain homeostasis, including the regulation of neurogenesis, cell survival, and synapse maturation and elimination, especially in the developing brain. In this review, we summarize the current state of knowledge on microglia in brain development, with a focus on their neuroprotective function. We will also discuss how microglial dysfunction may lead to the impairment of brain function, thereby contributing to disease development.
Collapse
|
8
|
Yuan Y, Wu C, Ling EA. Heterogeneity of Microglia Phenotypes: Developmental, Functional and Some Therapeutic Considerations. Curr Pharm Des 2020; 25:2375-2393. [PMID: 31584369 DOI: 10.2174/1381612825666190722114248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Microglia play a pivotal role in maintaining homeostasis in complex brain environment. They first exist as amoeboid microglial cells (AMCs) in the developing brain, but with brain maturation, they transform into ramified microglial cells (RMCs). In pathological conditions, microglia are activated and have been classified into M1 and M2 phenotypes. The roles of AMCs, RMCs and M1/M2 microglia phenotypes especially in pathological conditions have been the focus of many recent studies. METHODS Here, we review the early development of the AMCs and RMCs and discuss their specific functions with reference to their anatomic locations, immunochemical coding etc. M1 and M2 microglia phenotypes in different neuropathological conditions are also reviewed. RESULTS Activated microglia are engaged in phagocytosis, production of proinflammatory mediators, trophic factors and synaptogenesis etc. Prolonged microglia activation, however, can cause damage to neurons and oligodendrocytes. The M1 and M2 phenotypes featured prominently in pathological conditions are discussed in depth. Experimental evidence suggests that microglia phenotype is being modulated by multiple factors including external and internal stimuli, local demands, epigenetic regulation, and herbal compounds. CONCLUSION Prevailing views converge that M2 polarization is neuroprotective. Thus, proper therapeutic designs including the use of anti-inflammatory drugs, herbal agents may be beneficial in suppression of microglial activation, especially M1 phenotype, for amelioration of neuroinflammation in different neuropathological conditions. Finally, recent development of radioligands targeting 18 kDa translocator protein (TSPO) in activated microglia may hold great promises clinically for early detection of brain lesion with the positron emission tomography.
Collapse
Affiliation(s)
- Yun Yuan
- Department of Anatomy and Histology/Embryology, Kunming Medical University, 1168 West Chunrong Road, Kunming, China
| | - Chunyun Wu
- Department of Anatomy and Histology/Embryology, Kunming Medical University, 1168 West Chunrong Road, Kunming, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, 4 Medical Drive, MD10, National University of Singapore, 117594, Singapore
| |
Collapse
|
9
|
Rodríguez-Iglesias N, Sierra A, Valero J. Rewiring of Memory Circuits: Connecting Adult Newborn Neurons With the Help of Microglia. Front Cell Dev Biol 2019; 7:24. [PMID: 30891446 PMCID: PMC6411767 DOI: 10.3389/fcell.2019.00024] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/08/2019] [Indexed: 12/16/2022] Open
Abstract
New neurons are continuously generated from stem cells and integrated into the adult hippocampal circuitry, contributing to memory function. Several environmental, cellular, and molecular factors regulate the formation of new neurons, but the mechanisms that govern their incorporation into memory circuits are less explored. Herein we will focus on microglia, the resident immune cells of the CNS, which modulate the production of new neurons in the adult hippocampus and are also well suited to participate in their circuit integration. Microglia may contribute to the refinement of brain circuits during development and exert a role in physiological and pathological conditions by regulating axonal and dendritic growth; promoting the formation, elimination, and relocation of synapses; modulating excitatory synaptic maturation; and participating in functional synaptic plasticity. Importantly, microglia are able to sense subtle changes in their environment and may use this information to differently modulate hippocampal wiring, ultimately impacting on memory function. Deciphering the role of microglia in hippocampal circuitry constant rewiring will help to better understand the influence of microglia on memory function.
Collapse
Affiliation(s)
- Noelia Rodríguez-Iglesias
- Laboratory of Glial Cell Biology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Amanda Sierra
- Laboratory of Glial Cell Biology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Leioa, Spain.,Ikerbasque Foundation, Bilbao, Spain
| | - Jorge Valero
- Laboratory of Glial Cell Biology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Leioa, Spain.,Ikerbasque Foundation, Bilbao, Spain
| |
Collapse
|
10
|
Abstract
Microglia are neural cells of nonneural origin; they originate from fetal macrophages that invade neural tube early in embryogenesis and undergo the most idiosyncratic metamorphosis which coverts them into elements of neural circuitry. Microglia appeared early in evolution with neural immune cells being operative in leeches and mollusks. Microglial cells acquire specific morphology characterized by small cell bodies and long motile processes which are packed with receptors sensing both physiological and pathological stimuli. Microglial cells actively sculpture neuronal networks through synaptic stripping and phagocytosis of redundant neurons; microglia also secrete neuroactive factors regulating synaptic transmission. Novel techniques emerging in recent decade allowed an in-depth understanding of physiological and pathophysiological functions of microglia.
Collapse
Affiliation(s)
- Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
11
|
Konishi H, Kiyama H, Ueno M. Dual functions of microglia in the formation and refinement of neural circuits during development. Int J Dev Neurosci 2018; 77:18-25. [DOI: 10.1016/j.ijdevneu.2018.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/20/2018] [Accepted: 09/30/2018] [Indexed: 12/29/2022] Open
Affiliation(s)
- Hiroyuki Konishi
- Department of Functional Anatomy and NeuroscienceNagoya University Graduate School of MedicineNagoya466‐8550Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy and NeuroscienceNagoya University Graduate School of MedicineNagoya466‐8550Japan
| | - Masaki Ueno
- Department of System Pathology for Neurological DisordersBrain Research InstituteNiigata UniversityNiigata951‐8585Japan
| |
Collapse
|
12
|
Weinhard L, d'Errico P, Leng Tay T. Headmasters: Microglial regulation of learning and memory in health and disease. AIMS MOLECULAR SCIENCE 2018. [DOI: 10.3934/molsci.2018.1.63] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
13
|
Cantaut-Belarif Y, Antri M, Pizzarelli R, Colasse S, Vaccari I, Soares S, Renner M, Dallel R, Triller A, Bessis A. Microglia control the glycinergic but not the GABAergic synapses via prostaglandin E2 in the spinal cord. J Cell Biol 2017; 216:2979-2989. [PMID: 28716844 PMCID: PMC5584146 DOI: 10.1083/jcb.201607048] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 02/10/2017] [Accepted: 06/07/2017] [Indexed: 01/08/2023] Open
Abstract
Microglia can influence the excitatory responses of neurons, but less is known about how these immune cells in the brain may influence inhibitory neurotransmitters. Cantaut-Belarif et al. report that prostaglandin production by Toll-like receptor–stimulated microglia can influence the glycinergic but not GABAergic responses of neurons by altering the lateral diffusion of glycine receptors specifically within the synaptic membrane. Microglia control excitatory synapses, but their role in inhibitory neurotransmission has been less well characterized. Herein, we show that microglia control the strength of glycinergic but not GABAergic synapses via modulation of the diffusion dynamics and synaptic trapping of glycine (GlyR) but not GABAA receptors. We further demonstrate that microglia regulate the activity-dependent plasticity of glycinergic synapses by tuning the GlyR diffusion trap. This microglia–synapse cross talk requires production of prostaglandin E2 by microglia, leading to the activation of neuronal EP2 receptors and cyclic adenosine monophosphate–dependent protein kinase. Thus, we now provide a link between microglial activation and synaptic dysfunctions, which are common early features of many brain diseases.
Collapse
Affiliation(s)
- Yasmine Cantaut-Belarif
- École Normale Supérieure, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris Sciences et Lettres Research University, Paris, France
| | - Myriam Antri
- Faculté de Chirurgie Dentaire, Neuro-Dol, Centre Hospitalier Universitaire de Clermont-Ferrand, Université Clermont Auvergne, Institut National de la Santé et de la Recherche Médicale, Clermont-Ferrand, France
| | - Rocco Pizzarelli
- École Normale Supérieure, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris Sciences et Lettres Research University, Paris, France
| | - Sabrina Colasse
- École Normale Supérieure, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris Sciences et Lettres Research University, Paris, France
| | - Ilaria Vaccari
- École Normale Supérieure, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris Sciences et Lettres Research University, Paris, France
| | - Sylvia Soares
- Sorbonne Universités, UPMC, CNRS 8246, INSERM 1130, Institut de Biologie Paris-Seine, Neuroscience Paris Seine, Paris, France
| | - Marianne Renner
- École Normale Supérieure, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris Sciences et Lettres Research University, Paris, France
| | - Radhouane Dallel
- Faculté de Chirurgie Dentaire, Neuro-Dol, Centre Hospitalier Universitaire de Clermont-Ferrand, Université Clermont Auvergne, Institut National de la Santé et de la Recherche Médicale, Clermont-Ferrand, France
| | - Antoine Triller
- École Normale Supérieure, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris Sciences et Lettres Research University, Paris, France
| | - Alain Bessis
- École Normale Supérieure, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Paris Sciences et Lettres Research University, Paris, France
| |
Collapse
|
14
|
Ikawa D, Makinodan M, Iwata K, Ohgidani M, Kato TA, Yamashita Y, Yamamuro K, Kimoto S, Toritsuka M, Yamauchi T, Fukami SI, Yoshino H, Okumura K, Tanaka T, Wanaka A, Owada Y, Tsujii M, Sugiyama T, Tsuchiya K, Mori N, Hashimoto R, Matsuzaki H, Kanba S, Kishimoto T. Microglia-derived neuregulin expression in psychiatric disorders. Brain Behav Immun 2017; 61:375-385. [PMID: 28089559 DOI: 10.1016/j.bbi.2017.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 01/05/2017] [Accepted: 01/08/2017] [Indexed: 12/18/2022] Open
Abstract
Several studies have revealed that neuregulins (NRGs) are involved in brain function and psychiatric disorders. While NRGs have been regarded as neuron- or astrocyte-derived molecules, our research has revealed that microglia also express NRGs, levels of which are markedly increased in activated microglia. Previous studies have indicated that microglia are activated in the brains of individuals with autism spectrum disorder (ASD). Therefore, we investigated microglial NRG mRNA expression in multiple lines of mice considered models of ASD. Intriguingly, microglial NRG expression significantly increased in BTBR and socially-isolated mice, while maternal immune activation (MIA) mice exhibited identical NRG expression to controls. Furthermore, we observed a positive correlation between NRG expression in microglia and peripheral blood mononuclear cells (PBMCs) in mice, suggesting that NRG expression in human PBMCs may mirror microglia-derived NRG expression in the human brain. To translate these findings for application in clinical psychiatry, we measured levels of NRG1 splice-variant expression in clinically available PBMCs of patients with ASD. Levels of NRG1 type III expression in PBMCs were positively correlated with impairments in social interaction in children with ASD (as assessed using the Autistic Diagnostic Interview-Revised test: ADI-R). These findings suggest that immune cell-derived NRGs may be implicated in the pathobiology of psychiatric disorders such as ASD.
Collapse
Affiliation(s)
- Daisuke Ikawa
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan.
| | - Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, Japan; Department of Development of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Fukui, Japan
| | - Masahiro Ohgidani
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan; Innovation Center for Medical Redox Navigation, Kyushu University, Fukuoka, Japan
| | - Yasunori Yamashita
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Takahira Yamauchi
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Shin-Ichi Fukami
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Hiroki Yoshino
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Kazuki Okumura
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| | - Tatsuhide Tanaka
- Department of Anatomy and Neuroscience, Nara Medical University School of Medicine, Nara, Japan
| | - Akio Wanaka
- Department of Anatomy and Neuroscience, Nara Medical University School of Medicine, Nara, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | | | | | - Kenji Tsuchiya
- Department of Psychiatry and Neurology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Norio Mori
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan
| | - Ryota Hashimoto
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan; Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui, Japan; Department of Development of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Fukui, Japan
| | - Shigenobu Kanba
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University School of Medicine, Nara, Japan
| |
Collapse
|
15
|
Mosser CA, Baptista S, Arnoux I, Audinat E. Microglia in CNS development: Shaping the brain for the future. Prog Neurobiol 2017; 149-150:1-20. [DOI: 10.1016/j.pneurobio.2017.01.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 12/22/2022]
|
16
|
Reemst K, Noctor SC, Lucassen PJ, Hol EM. The Indispensable Roles of Microglia and Astrocytes during Brain Development. Front Hum Neurosci 2016; 10:566. [PMID: 27877121 PMCID: PMC5099170 DOI: 10.3389/fnhum.2016.00566] [Citation(s) in RCA: 357] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 10/25/2016] [Indexed: 01/17/2023] Open
Abstract
Glia are essential for brain functioning during development and in the adult brain. Here, we discuss the various roles of both microglia and astrocytes, and their interactions during brain development. Although both cells are fundamentally different in origin and function, they often affect the same developmental processes such as neuro-/gliogenesis, angiogenesis, axonal outgrowth, synaptogenesis and synaptic pruning. Due to their important instructive roles in these processes, dysfunction of microglia or astrocytes during brain development could contribute to neurodevelopmental disorders and potentially even late-onset neuropathology. A better understanding of the origin, differentiation process and developmental functions of microglia and astrocytes will help to fully appreciate their role both in the developing as well as in the adult brain, in health and disease.
Collapse
Affiliation(s)
- Kitty Reemst
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| | - Stephen C. Noctor
- Department of Psychiatry and Behavioral Sciences, UC Davis MIND InstituteSacramento, CA, USA
| | - Paul J. Lucassen
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| | - Elly M. Hol
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
- Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands
- Netherlands Institute for NeuroscienceAmsterdam, Netherlands
| |
Collapse
|
17
|
von Bernhardi R, Heredia F, Salgado N, Muñoz P. Microglia Function in the Normal Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:67-92. [PMID: 27714685 DOI: 10.1007/978-3-319-40764-7_4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The activation of microglia has been recognized for over a century by their morphological changes. Long slender microglia acquire a short sturdy ramified shape when activated. During the past 20 years, microglia have been accepted as an essential cellular component for understanding the pathogenic mechanism of many brain diseases, including neurodegenerative diseases. More recently, functional studies and imaging in mouse models indicate that microglia are active in the healthy central nervous system. It has become evident that microglia release several signal molecules that play key roles in the crosstalk among brain cells, i.e., astrocytes and oligodendrocytes with neurons, as well as with regulatory immune cells. Recent studies also reveal the heterogeneous nature of microglia diverse functions depending on development, previous exposure to stimulation events, brain region of residence, or pathological state. Subjects to approach by future research are still the unresolved questions regarding the conditions and mechanisms that render microglia protective, capable of preventing or reducing damage, or deleterious, capable of inducing or facilitating the progression of neuropathological diseases. This novel knowledge will certainly change our view on microglia as therapeutic target, shifting our goal from their general silencing to the generation of treatments able to change their activation pattern.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile.
| | - Florencia Heredia
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Nicole Salgado
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Paola Muñoz
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| |
Collapse
|
18
|
Risher ML, Sexton HG, Risher WC, Wilson WA, Fleming RL, Madison RD, Moore SD, Eroglu C, Swartzwelder HS. Adolescent Intermittent Alcohol Exposure: Dysregulation of Thrombospondins and Synapse Formation are Associated with Decreased Neuronal Density in the Adult Hippocampus. Alcohol Clin Exp Res 2015; 39:2403-13. [PMID: 26537975 PMCID: PMC4712076 DOI: 10.1111/acer.12913] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/22/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND Adolescent intermittent alcohol exposure (AIE) has profound effects on neuronal function. We have previously shown that AIE causes aberrant hippocampal structure and function that persists into adulthood. However, the possible contributions of astrocytes and their signaling factors remain largely unexplored. We investigated the acute and enduring effects of AIE on astrocytic reactivity and signaling on synaptic expression in the hippocampus, including the impact of the thrombospondin (TSP) family of astrocyte-secreted synaptogenic factors and their neuronal receptor, alpha2delta-1 (α2δ-1). Our hypothesis is that some of the influences of AIE on neuronal function may be secondary to direct effects on astrocytes. METHODS We conducted Western blot analysis on TSPs 1 to 4 and α2δ-1 from whole hippocampal lysates 24 hours after the 4th and 10th doses of AIE, then 24 days after the last dose (in adulthood). We used immunohistochemistry to assess astrocyte reactivity (i.e., morphology) and synaptogenesis (i.e., colocalization of pre- and postsynaptic puncta). RESULTS Adolescent AIE reduced α2δ-1 expression, and colocalized pre- and postsynaptic puncta after the fourth ethanol (EtOH) dose. By the 10th dose, increased TSP2 levels were accompanied by an increase in colocalized pre- and postsynaptic puncta, while α2δ-1 returned to control levels. Twenty-four days after the last EtOH dose (i.e., adulthood), TSP2, TSP4, and α2δ-1 expression were all elevated. Astrocyte reactivity, indicated by increased astrocytic volume and area, was also observed at that time. CONCLUSIONS Repeated EtOH exposure during adolescence results in long-term changes in specific astrocyte signaling proteins and their neuronal synaptogenic receptor. Continued signaling by these traditionally developmental factors in adulthood may represent a compensatory mechanism whereby astrocytes reopen the synaptogenic window and repair lost connectivity, and consequently contribute to the enduring maladaptive structural and functional abnormalities previously observed in the hippocampus after AIE.
Collapse
Affiliation(s)
- Mary-Louise Risher
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - Hannah G Sexton
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - W Christopher Risher
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| | - Wilkie A Wilson
- Social Sciences Research Institute, Duke University Medical Center, Durham, North Carolina
| | - Rebekah L Fleming
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - Roger D Madison
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Scott D Moore
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| | - H Scott Swartzwelder
- Durham VA Medical Center, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Department of Psychology and Neuroscience, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
19
|
Delpech JC, Madore C, Nadjar A, Joffre C, Wohleb ES, Layé S. Microglia in neuronal plasticity: Influence of stress. Neuropharmacology 2015; 96:19-28. [PMID: 25582288 DOI: 10.1016/j.neuropharm.2014.12.034] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/24/2014] [Accepted: 12/29/2014] [Indexed: 01/17/2023]
Abstract
The central nervous system (CNS) has previously been regarded as an immune-privileged site with the absence of immune cell responses but this dogma was not entirely true. Microglia are the brain innate immune cells and recent findings indicate that they participate both in CNS disease and infection as well as facilitate normal CNS function. Microglia are highly plastic and play integral roles in sculpting the structure of the CNS, refining neuronal circuitry and connectivity, and contribute actively to neuronal plasticity in the healthy brain. Interestingly, psychological stress can perturb the function of microglia in association with an impaired neuronal plasticity and the development of emotional behavior alterations. As a result it seemed important to describe in this review some findings indicating that the stress-induced microglia dysfunction may underlie neuroplasticity deficits associated to many mood disorders. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Jean-Christophe Delpech
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France
| | - Charlotte Madore
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France
| | - Agnes Nadjar
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France
| | - Corinne Joffre
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France
| | - Eric S Wohleb
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Sophie Layé
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France.
| |
Collapse
|
20
|
Glial cells as key players in schizophrenia pathology: recent insights and concepts of therapy. Schizophr Res 2015; 161:4-18. [PMID: 24948484 DOI: 10.1016/j.schres.2014.03.035] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/27/2014] [Accepted: 03/01/2014] [Indexed: 02/07/2023]
Abstract
The past decade has witnessed an explosion of knowledge on the impact of glia for the neurobiological foundation of schizophrenia. A plethora of studies have shown structural and functional abnormalities in all three types of glial cells. There is convincing evidence of reduced numbers of oligodendrocytes, impaired cell maturation and altered gene expression of myelin/oligodendrocyte-related genes that may in part explain white matter abnormalities and disturbed inter- and intra-hemispheric connectivity, which are characteristic signs of schizophrenia. Earlier reports of astrogliosis could not be confirmed by later studies, although the expression of a variety of astrocyte-related genes is abnormal in psychosis. Since astrocytes play a key role in the synaptic metabolism of glutamate, GABA, monoamines and purines, astrocyte dysfunction may contribute to certain aspects of disturbed neurotransmission in schizophrenia. Finally, increased densities of microglial cells and aberrant expression of microglia-related surface markers in schizophrenia suggest that immunological/inflammatory factors are of considerable relevance for the pathophysiology of psychosis. This review describes current evidence for the multifaceted role of glial cells in schizophrenia and discusses efforts to develop glia-directed therapies for the treatment of the disease.
Collapse
|
21
|
Nieto-Rostro M, Sandhu G, Bauer CS, Jiruska P, Jefferys JGR, Dolphin AC. Altered expression of the voltage-gated calcium channel subunit α₂δ-1: a comparison between two experimental models of epilepsy and a sensory nerve ligation model of neuropathic pain. Neuroscience 2014; 283:124-37. [PMID: 24641886 PMCID: PMC4259901 DOI: 10.1016/j.neuroscience.2014.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/03/2014] [Accepted: 03/09/2014] [Indexed: 12/20/2022]
Abstract
The auxiliary α2δ-1 subunit of voltage-gated calcium channels is up-regulated in dorsal root ganglion neurons following peripheral somatosensory nerve damage, in several animal models of neuropathic pain. The α2δ-1 protein has a mainly presynaptic localization, where it is associated with the calcium channels involved in neurotransmitter release. Relevant to the present study, α2δ-1 has been shown to be the therapeutic target of the gabapentinoid drugs in their alleviation of neuropathic pain. These drugs are also used in the treatment of certain epilepsies. In this study we therefore examined whether the level or distribution of α2δ-1 was altered in the hippocampus following experimental induction of epileptic seizures in rats, using both the kainic acid model of human temporal lobe epilepsy, in which status epilepticus is induced, and the tetanus toxin model in which status epilepticus is not involved. The main finding of this study is that we did not identify somatic overexpression of α2δ-1 in hippocampal neurons in either of the epilepsy models, unlike the upregulation of α2δ-1 that occurs following peripheral nerve damage to both somatosensory and motor neurons. However, we did observe local reorganization of α2δ-1 immunostaining in the hippocampus only in the kainic acid model, where it was associated with areas of neuronal cell loss, as indicated by absence of NeuN immunostaining, dendritic loss, as identified by areas where microtubule-associated protein-2 immunostaining was missing, and reactive gliosis, determined by regions of strong OX42 staining.
Collapse
Affiliation(s)
- M Nieto-Rostro
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - G Sandhu
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - C S Bauer
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - P Jiruska
- Neuronal Networks Group, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, UK
| | - J G R Jefferys
- Neuronal Networks Group, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, UK
| | - A C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
22
|
Ganguly P, Brenhouse HC. Broken or maladaptive? Altered trajectories in neuroinflammation and behavior after early life adversity. Dev Cogn Neurosci 2014; 11:18-30. [PMID: 25081071 PMCID: PMC4476268 DOI: 10.1016/j.dcn.2014.07.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/23/2014] [Accepted: 07/02/2014] [Indexed: 12/11/2022] Open
Abstract
This paper reviews how early life adversity alters neuroimmune mechanisms. Neuroimmune sensitization from early life adversity impacts circuitry at discrete life stages. Neuroimmune and neurodevelopmental influences can impact behavior and vulnerability. Sexual dimorphism in immune and brain development yield distinct effects of early life adversity.
Exposure to adversity and stress early in development yields vulnerability to mental illnesses throughout the lifespan. Growing evidence suggests that this vulnerability has mechanistic origins involving aberrant development of both neurocircuitry and neuro-immune activity. Here we review the current understanding of when and how stress exposure initiates neuroinflammatory events that interact with brain development. We first review how early life adversity has been associated with various psychopathologies, and how neuroinflammation plays a role in these pathologies. We then summarize data and resultant hypotheses describing how early life adversity may particularly alter neuro-immune development with psychiatric consequences. Finally, we review how sex differences contribute to individualistic vulnerabilities across the lifespan. We submit the importance of understanding how stress during early development might cause outright neural or glial damage, as well as experience-dependent plasticity that may insufficiently prepare an individual for sex-specific or life-stage specific challenges.
Collapse
Affiliation(s)
- Prabarna Ganguly
- Northeastern University, Psychology Department, 125 Nightingale Hall, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Heather C Brenhouse
- Northeastern University, Psychology Department, 125 Nightingale Hall, 360 Huntington Avenue, Boston, MA 02115, USA.
| |
Collapse
|
23
|
Ueno M, Yamashita T. Bidirectional tuning of microglia in the developing brain: from neurogenesis to neural circuit formation. Curr Opin Neurobiol 2014; 27:8-15. [PMID: 24607651 DOI: 10.1016/j.conb.2014.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/15/2014] [Accepted: 02/06/2014] [Indexed: 12/14/2022]
Abstract
The developing brain employs multi-step processes to construct neural circuitry. Recent studies have highlighted that microglia, traditionally known to be the resident immune cells in the brain, have essential roles in these processes, which range from neurogenesis to establishing synaptic connections. Microglia play bidirectional roles for maintaining proper circuitry: eliminating unnecessary cells, axons, and synapses, while supporting the neighboring ones. Although these processes are performed in different parts of the neuron, similar molecular mechanisms are possibly involved. This paper reviews recent progress on the knowledge of the roles of microglia in brain development, and further discusses the application of this knowledge in therapies for brain disorders and injuries.
Collapse
Affiliation(s)
- Masaki Ueno
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, United States; Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita-shi, Osaka 565-0871, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 5 Sanbancho, Chiyoda-ku, Tokyo, Japan.
| |
Collapse
|
24
|
Blaylock RL. Immunology primer for neurosurgeons and neurologists part 2: Innate brain immunity. Surg Neurol Int 2013; 4:118. [PMID: 24083053 PMCID: PMC3784951 DOI: 10.4103/2152-7806.118349] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 06/18/2013] [Indexed: 12/27/2022] Open
Abstract
Over the past several decades we have learned a great deal about microglia and innate brain immunity. While microglia are the principle innate immune cells, other cell types also play a role, including invading macrophages, astrocytes, neurons, and endothelial cells. The fastest reacting cell is the microglia and despite its name, resting microglia (also called ramified microglia) are in fact quite active. Motion photomicrographs demonstrate a constant movement of ramified microglial foot processes, which appear to be testing the microenvironment for dangerous alteration in extracellular fluid content. These foot processes, in particular, interact with synapses and play a role in synaptic function. In event of excitatory overactivity, these foot processes can strip selected synapses, thus reducing activation states as a neuroprotective mechanism. They can also clear extracellular glutamate so as to reduce the risk of excitotoxicity. Microglia also appear to have a number of activation phenotypes, such as: (1) phagocytic, (2) neuroprotective and growth promoting, or (3) primarily neurodestructive. These innate immune cells can migrate a great distance under pathological conditions and appear to have anatomic specificity, meaning they can accumulate in specifically selected areas of the brain. There is some evidence that there are several types of microglia. Macrophage infiltration into the embryonic brain is the source of resident microglia and in adulthood macrophages can infiltrate the brain and are for the most part pathologically indistinguishable from resident microglia, but may react differently. Activation itself does not imply a destructive phenotype and can be mostly neuroprotective via phagocytosis of debris, neuron parts and dying cells and by the release of neurotrophins such as nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF). Evidence is accumulating that microglia undergo dynamic fluctuations in phenotype as the neuropathology evolves. For example, in the early stages of neurotrauma and stroke, microglia play a mostly neuroprotective role and only later switch to a neurodestructive mode. A great number of biological systems alter microglia function, including neurohormones, cannabinoids, other neurotransmitters, adenosine triphosphate (ATP), adenosine, and corticosteroids. One can appreciate that with aging many of these systems are altered by the aging process itself or by disease thus changing the sensitivity of the innate immune system.
Collapse
Affiliation(s)
- Russell L Blaylock
- Theoretical Neurosciences Research, LLC, Neurosurgeon (Ret), Ridgeland, MS
| |
Collapse
|
25
|
Beumer W, Gibney SM, Drexhage RC, Pont-Lezica L, Doorduin J, Klein HC, Steiner J, Connor TJ, Harkin A, Versnel MA, Drexhage HA. The immune theory of psychiatric diseases: a key role for activated microglia and circulating monocytes. J Leukoc Biol 2012; 92:959-75. [PMID: 22875882 DOI: 10.1189/jlb.0212100] [Citation(s) in RCA: 277] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
This review describes a key role for mononuclear phagocytes in the pathogenesis of major psychiatric disorders. There is accumulating evidence for activation of microglia (histopathology and PET scans) and circulating monocytes (enhanced gene expression of immune genes, an overproduction of monocyte/macrophage-related cytokines) in patients with bipolar disorder, major depressive disorder, and schizophrenia. These data are strengthened by observations in animal models, such as the MIA models, the chronic stress models, and the NOD mouse model. In these animal models of depressive-, anxiety-, and schizophrenia-like behavior, similar activations of microglia and circulating monocytes can be found. These animal models also make in-depth pathogenic studies possible and show that microglia activation impacts neuronal development and function in brain areas congruent with the altered depressive and schizophrenia-like behaviors.
Collapse
Affiliation(s)
- Wouter Beumer
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Autism spectrum disorders (ASDs) including classic autism is a group of complex developmental disabilities with core deficits of impaired social interactions, communication difficulties and repetitive behaviors. Although the neurobiology of ASDs has attracted much attention in the last two decades, the role of microglia has been ignored. Existing data are focused on their recognized role in neuroinflammation, which only covers a small part of the pathological repertoire of microglia. This review highlights recent findings on the broader roles of microglia, including their active surveillance of brain microenvironments and regulation of synaptic connectivity, maturation of brain circuitry and neurogenesis. Emerging evidence suggests that microglia respond to pre- and postnatal environmental stimuli through epigenetic interface to change gene expression, thus acting as effectors of experience-dependent synaptic plasticity. Impairments of these microglial functions could substantially contribute to several major etiological factors of autism, such as environmental toxins and cortical underconnectivity. Our recent study on Rett syndrome, a syndromic autistic disorder, provides an example that intrinsic microglial dysfunction due to genetic and epigenetic aberrations could detrimentally affect the developmental trajectory without evoking neuroinflammation. We propose that ASDs provide excellent opportunities to study the influence of microglia on neurodevelopment, and this knowledge could lead to novel therapies.
Collapse
|
27
|
Abstract
Microglial cells are the resident macrophages in the central nervous system. These cells of mesodermal/mesenchymal origin migrate into all regions of the central nervous system, disseminate through the brain parenchyma, and acquire a specific ramified morphological phenotype termed "resting microglia." Recent studies indicate that even in the normal brain, microglia have highly motile processes by which they scan their territorial domains. By a large number of signaling pathways they can communicate with macroglial cells and neurons and with cells of the immune system. Likewise, microglial cells express receptors classically described for brain-specific communication such as neurotransmitter receptors and those first discovered as immune cell-specific such as for cytokines. Microglial cells are considered the most susceptible sensors of brain pathology. Upon any detection of signs for brain lesions or nervous system dysfunction, microglial cells undergo a complex, multistage activation process that converts them into the "activated microglial cell." This cell form has the capacity to release a large number of substances that can act detrimental or beneficial for the surrounding cells. Activated microglial cells can migrate to the site of injury, proliferate, and phagocytose cells and cellular compartments.
Collapse
|
28
|
Verney C, Monier A, Fallet-Bianco C, Gressens P. Early microglial colonization of the human forebrain and possible involvement in periventricular white-matter injury of preterm infants. J Anat 2011; 217:436-48. [PMID: 20557401 DOI: 10.1111/j.1469-7580.2010.01245.x] [Citation(s) in RCA: 174] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Amoeboid microglial subpopulations visualized by antibodies against ionized calcium-binding adapter molecule 1, CD68, and CD45 enter the forebrain starting at 4.5 postovulatory or gestational weeks (gw). They penetrate the telencephalon and diencephalon via the meninges, choroid plexus, and ventricular zone. Early colonization by amoeboid microglia-macrophages is first restricted to the white matter, where these cells migrate and accumulate in patches at the junctions of white-matter pathways, such as the three junctions that the internal capsule makes with the thalamocortical projection, external capsule and cerebral peduncle, respectively. In the cerebral cortex anlage, migration is mainly radial and tangential towards the immature white matter, subplate layer, and cortical plate, whereas pial cells populate the prospective layer I. A second wave of microglial cells penetrates the brain via the vascular route at about 12-13 gw and remains confined to the white matter. Two main findings deserve emphasis. First, microglia accumulate at 10-12 gw at the cortical plate-subplate junction, where the first synapses are detected. Second, microglia accumulate in restricted laminar bands, most notably around 19-30 gw, at the axonal crossroads in the white matter (semiovale centre) rostrally, extending caudally in the immature white matter to the visual radiations. This accumulation of proliferating microglia is located at the site of white-matter injury in premature neonates. The spatiotemporal organization of microglia in the immature white and grey matter suggests that these cells may play active roles in developmental processes such as axonal guidance, synaptogenesis, and neurodevelopmental apoptosis as well as in injuries to the developing brain, in particular in the periventricular white-matter injury of preterm infants.
Collapse
Affiliation(s)
- Catherine Verney
- INSERM U676, Hôpital Robert Debré, 48 Boulevard Sérurier, Paris, France
| | | | | | | |
Collapse
|
29
|
Rigato C, Buckinx R, Le-Corronc H, Rigo JM, Legendre P. Pattern of invasion of the embryonic mouse spinal cord by microglial cells at the time of the onset of functional neuronal networks. Glia 2011; 59:675-95. [PMID: 21305616 DOI: 10.1002/glia.21140] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 12/15/2010] [Indexed: 01/30/2023]
Abstract
Microglial cells invade the central nervous system during embryonic development, but their developmental functional roles in vivo remain largely unknown. Accordingly, their invasion pattern during early embryonic development is still poorly understood. To address this issue, we analyzed the initial developmental pattern of microglial cell invasion in the spinal cord of CX3CR1-eGFP mouse embryos using immunohistochemistry. Microglial cells began to invade the mouse embryonic spinal cord at a developmental period corresponding to the onset of spontaneous electrical activity and of synaptogenesis. Microglial cells reached the spinal cord through the peripheral vasculature and began to invade the parenchyma at 11.5 days of embryonic age (E11.5). Remarkably, at E12.5, activated microglial cells aggregated in the dorsolateral region close to terminals of dying dorsal root ganglia neurons. At E13.5, microglial cells in the ventral marginal zone interacted with radial glial cells, whereas ramified microglial cells within the parenchyma interacted with growing capillaries. At this age, activated microglial cells (Mac-2 staining) also accumulated within the lateral motor columns at the onset of the developmental cell death of motoneurons. This cell aggregation was still observed at E14.5, but microglial cells no longer expressed Mac-2. At E15.5, microglial cells were randomly distributed within the parenchyma. Our results provide the essential basis for further studies on the role of microglial cells in the early development of spinal cord neuronal networks in vivo.
Collapse
Affiliation(s)
- C Rigato
- Institut National de la Santé et de la Recherche Médicale, U952, Université Pierre et Marie Curie, Paris, Ile de France, France
| | | | | | | | | |
Collapse
|
30
|
Effect of Exposure to 1,800 MHz Electromagnetic Fields on Heat Shock Proteins and Glial Cells in the Brain of Developing Rats. Neurotox Res 2010; 20:109-19. [DOI: 10.1007/s12640-010-9225-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 10/04/2010] [Accepted: 10/05/2010] [Indexed: 09/29/2022]
|
31
|
Chaves-da-Silva PG, de Barros CM, Lima FRS, Biancalana A, Martinez AMB, Allodi S. Identity of the cells recruited to a lesion in the central nervous system of a decapod crustacean. Cell Tissue Res 2010; 342:179-89. [DOI: 10.1007/s00441-010-1045-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 09/01/2010] [Indexed: 10/19/2022]
|
32
|
Drexhage RC, Knijff EM, Padmos RC, Heul-Nieuwenhuijzen LVD, Beumer W, Versnel MA, Drexhage HA. The mononuclear phagocyte system and its cytokine inflammatory networks in schizophrenia and bipolar disorder. Expert Rev Neurother 2010; 10:59-76. [PMID: 20021321 DOI: 10.1586/ern.09.144] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This review describes patients with schizophrenia and bipolar disorder. In such patients, a high inflammatory set point of circulating monocytes at the transcriptome level is observed, involving various inflammatory transcripts forming distinct fingerprints (the transcriptomic monocyte fingerprint in schizophrenia overlaps with that in bipolar disorder, but also differs with it at points). There are increased levels of compounds of the IL-1, IL-6 and TNF system in the serum (be it modest and inconsistent). There is also evidence that the IL-2 system is activated in patients with schizophrenia (and perhaps those with mania), although independently of the activation of the IL-1, IL-6 and TNF systems, suggesting separate inducing mechanisms for monocyte and T-cell activation. It is not yet known whether such T cell activation involves the Th1/Th2/Th17 or Treg systems.
Collapse
Affiliation(s)
- Roosmarijn C Drexhage
- Department of Immunology, Erasmus MC, PO Box 2040, 3000CA Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
33
|
Madinier A, Bertrand N, Mossiat C, Prigent-Tessier A, Beley A, Marie C, Garnier P. Microglial involvement in neuroplastic changes following focal brain ischemia in rats. PLoS One 2009; 4:e8101. [PMID: 19956568 PMCID: PMC2779656 DOI: 10.1371/journal.pone.0008101] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 11/05/2009] [Indexed: 12/11/2022] Open
Abstract
The pathogenesis of ischemic stroke is a complex sequence of events including inflammatory reaction, for which the microglia appears to be a major cellular contributor. However, whether post-ischemic activation of microglial cells has beneficial or detrimental effects remains to be elucidated, in particular on long term brain plasticity events. The objective of our study was to determine, through modulation of post-stroke inflammatory response, to what extent microglial cells are involved in some specific events of neuronal plasticity, neurite outgrowth and synaptogenesis. Since microglia is a source of neurotrophic factors, the identification of the brain-derived neurophic factor (BDNF) as possible molecular actor involved in these events was also attempted. As a means of down-regulating the microglial response induced by ischemia, 3-aminobenzamide (3-AB, 90 mg/kg, i.p.) was used to inhibit the poly(ADP-ribose) polymerase-1 (PARP-1). Indeed, PARP-1 contributes to the activation of the transcription factor NF-kB, which is essential to the upregulation of proinflammatory genes, in particular responsible for microglial activation/proliferation. Experiments were conducted in rats subjected to photothrombotic ischemia which leads to a strong and early microglial cells activation/proliferation followed by an infiltration of macrophages within the cortical lesion, events evaluated at serial time points up to 1 month post-ictus by immunostaining for OX-42 and ED-1. Our most striking finding was that the decrease in acute microglial activation induced by 3-AB was associated with a long term down-regulation of two neuronal plasticity proteins expression, synaptophysin (marker of synaptogenesis) and GAP-43 (marker of neuritogenesis) as well as to a significant decrease in tissue BDNF production. Thus, our data argue in favour of a supportive role for microglia in brain neuroplasticity stimulation possibly through BDNF production, suggesting that a targeted protection of microglial cells could represent an innovative approach to potentiate post-stroke neuroregeneration.
Collapse
Affiliation(s)
- Alexandre Madinier
- Unité INSERM U887 Motricité-Plasticité, Dijon, France
- Université de Bourgogne, Dijon, France
| | - Nathalie Bertrand
- Unité INSERM U887 Motricité-Plasticité, Dijon, France
- Université de Bourgogne, Dijon, France
- Département Génie Biologique, IUT, Dijon, France
| | - Claude Mossiat
- Unité INSERM U887 Motricité-Plasticité, Dijon, France
- Université de Bourgogne, Dijon, France
| | - Anne Prigent-Tessier
- Unité INSERM U887 Motricité-Plasticité, Dijon, France
- Université de Bourgogne, Dijon, France
| | - Alain Beley
- Université de Bourgogne, Dijon, France
- Département Génie Biologique, IUT, Dijon, France
| | - Christine Marie
- Unité INSERM U887 Motricité-Plasticité, Dijon, France
- Université de Bourgogne, Dijon, France
| | - Philippe Garnier
- Unité INSERM U887 Motricité-Plasticité, Dijon, France
- Université de Bourgogne, Dijon, France
- Département Génie Biologique, IUT, Dijon, France
- * E-mail:
| |
Collapse
|
34
|
Jonakait GM. The effects of maternal inflammation on neuronal development: possible mechanisms. Int J Dev Neurosci 2007; 25:415-25. [DOI: 10.1016/j.ijdevneu.2007.08.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Accepted: 08/27/2007] [Indexed: 10/22/2022] Open
Affiliation(s)
- G. Miller Jonakait
- Department of Biological SciencesNew Jersey Institute of Technology195 University AvenueNewarkNJ07102United States
| |
Collapse
|
35
|
Kaur C, Dheen ST, Ling EA. From blood to brain: amoeboid microglial cell, a nascent macrophage and its functions in developing brain. Acta Pharmacol Sin 2007; 28:1087-96. [PMID: 17640468 DOI: 10.1111/j.1745-7254.2007.00625.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Amoeboid microglial cells (AMC) in the developing brain are active macrophages. The macrophagic nature of these cells has been demonstrated by many methods, such as the localization of various hydrolytic enzymes and the presence of complement type 3 surface receptors in them. More importantly is the direct visualization of these cells engaged in the phagocytosis of degenerating cells at the ultrastructural level. Further evidence of them being active macrophages is the avid internalization of tracers administered by the intravenous or intraperitoneal routes in developing rats. The potential involvement of AMC in immune functions is supported by the induced expression of major histocompatibility complex class I and II antigens on them when challenged by lipopolysaccharide or interferon-gamma. Immunosuppressive drugs, such as glucocorticoids and immune function-enhancing drugs like melatonin, affect the expression of surface receptors and antigens and the release of cytokines by AMC. Recent studies in our laboratory have shown the expression of insulin-like growth factors, endothelins, 2',3'-cyclic nucleotide 3'-phosphodiesterase, and N-methyl-D-asparate receptors. This along with the release of chemokines, such as stromal derived factor-1a and monocyte chemoattractant protein-1, suggests multiple functional roles of AMC in early brain development.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | | | | |
Collapse
|
36
|
Rasouli A, Bhatia N, Suryadevara S, Cahill K, Gupta R. Transplantation of preconditioned schwann cells in peripheral nerve grafts after contusion in the adult spinal cord. Improvement of recovery in a rat model. J Bone Joint Surg Am 2006; 88:2400-10. [PMID: 17079397 DOI: 10.2106/jbjs.e.01424] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Recovery after injury to the peripheral nervous system is based on the pro-regenerative relationship between axons and the extracellular matrix, a relationship established by Schwann cells. As mechanical conditioning of Schwann cells has been shown to stimulate their regenerative behavior, we sought to determine whether transplantation of these cells to the central nervous system (i.e., the spinal cord), with its limited regenerative capacity after injury, would improve axonal regeneration and functional recovery. METHODS A moderate contusion injury of the spinal cord was created with a force-directed impactor in forty-eight adult Sprague-Dawley rats, and, at one week postinjury, the spinal cords were reexposed in all animals. In twenty-four of these animals, peripheral nerve grafts with Schwann cells that had been obtained from the sciatic nerves of donor animals, and had been either untreated or subjected to mechanical conditioning, were transplanted to the contused area of the cords following resection of the glial scar. Another group of animals was treated with glial scar excision only, and a fourth group had the contusion injury but neither glial excision nor transplantation. Scores according to the Basso, Beattie, Bresnahan (BBB) Locomotor Rating Scale were assigned preoperatively and weekly thereafter. Tract tracing of descending and ascending spinal cord tracts was performed at six weeks postoperatively for quantitative histological evaluation of axonal regeneration. RESULTS While the recovery following glial scar excision without peripheral nerve transplantation was significantly worse than the recovery in the other groups, both transplantation groups had significantly higher BBB scores than the controls (no transplantation) in the early postoperative period (p < 0.05). Moreover, histological analysis showed markedly increased axonal regeneration at the lesional sites in the animals treated with the mechanically conditioned grafts than in the other groups (p < 0.05). CONCLUSIONS Functional recovery after spinal cord contusion improved following glial scar excision with transplantation of Schwann cells in peripheral nerve grafts to the contusion areas. Although recovery did not differ significantly between the transplantation groups, only the preconditioned grafts led to axonal regeneration at and past the lesional site. These grafts may further enhance functional recovery as the descending tracts eventually reach their target end-organs.
Collapse
Affiliation(s)
- Alexandre Rasouli
- University of California, Irvine, 2226 Gillespie Neuroscience Research Facility, Irvine, CA 92697, USA
| | | | | | | | | |
Collapse
|
37
|
Monier A, Evrard P, Gressens P, Verney C. Distribution and differentiation of microglia in the human encephalon during the first two trimesters of gestation. J Comp Neurol 2006; 499:565-82. [PMID: 17029271 DOI: 10.1002/cne.21123] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We describe the topographical distribution of microglial subpopulations during development of the human diencephalon and telencephalon. Brains from embryos and fetuses age 5-23.5 gestational weeks (gw) were subjected to single- and double-immunolabeling for lectin RCA-1 (Ricinus Communis Agglutinin 1), Iba1 (a microglial marker), CD68 (specific of macrophages), CD45 (marker for mononucleate cells of hematopoietic lineage), CD34 (expressed on endothelial cells), and MIB1 and Ki67 (markers for cell proliferation). At 5.5 gw the first intracerebral microglial cells were seen close to the meninges and choroid plexus near the di-telencephalic fissure. They were amoeboid and positive for Iba1, CD45, and RCA-1, whereas cells in the deep parenchyma expressed Iba1/CD68/RCA-1 and constituted clusters. In the developing diencephalon, microglial clusters were located in junctional regions of the white matter anlagen, most notably at the junctions of the internal capsule with the thalamic projections, the external capsule, and the cerebral peduncle. In the cortical anlagen, Iba1+/RCA-1/CD68+/CD45+ cells accumulated at 10-12 gw, constituting a tangential band at the junction between the cortical plate and the subplate. Between 10 and 16 gw microglial clusters increased markedly in size and cellular density. Contact between Iba1+ microglia and CD34+ blood vessels was clearly visible from 10-12 gw onward, first in microglial clusters of the white matter anlagen and subsequently throughout the parenchyma. From the middle of the second trimester onward microglial cells colonized the entire cerebral parenchyma, developed a ramified morphology, and downregulated their surface antigens, but remained more numerous in the white matter.
Collapse
|
38
|
Bessis A, Béchade C, Bernard D, Roumier A. Microglial control of neuronal death and synaptic properties. Glia 2006; 55:233-8. [PMID: 17106878 DOI: 10.1002/glia.20459] [Citation(s) in RCA: 340] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Microglia have long been characterized by their immune function in the nervous system and are still mainly considered in a beneficial versus detrimental dialectic. However a review of literature enables to shed novel lights on microglial function under physiological conditions. It is now relevant to position these cells as full time partners of neuronal function and more specifically of synaptogenesis and developmental apoptosis. Indeed, microglia can actively control neuronal death. It has actually been shown in retina that microglial nerve growth factor (NGF) is necessary for the developmental apoptosis to occur. Similarly, in cerebellum, microglia induces developmental Purkinje cells death through respiratory burst. Furthermore, in spinal cord, microglial TNFalpha commits motoneurons to a neurotrophic dependent developmental apoptosis. Microglia can also control synaptogenesis. This is suggested by the fact that a mutation in KARAP/DAP12, a key protein of microglial activation impacts synaptic functions in hippocampus, and synapses protein content. In addition it has been now demonstrated that microglial brain-derived neurotrophin factor (BDNF) directly regulates synaptic properties in spinal cord. In conclusion, microglia can control neuronal function under physiological conditions and it is known that neuronal activity reciprocally controls microglial activation. We will discuss the importance of this cross-talk which allows microglia to orchestrate the balance between synaptogenesis and neuronal death occurring during development or injuries.
Collapse
Affiliation(s)
- Alain Bessis
- Biologie Cellulaire de la Synapse, Inserm U789, Ecole Normale Supérieure, 46 rue d'Ulm 75005 Paris, France.
| | | | | | | |
Collapse
|
39
|
Huleihel M, Golan H, Hallak M. Intrauterine infection/inflammation during pregnancy and offspring brain damages: possible mechanisms involved. Reprod Biol Endocrinol 2004; 2:17. [PMID: 15104793 PMCID: PMC411057 DOI: 10.1186/1477-7827-2-17] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2004] [Accepted: 04/22/2004] [Indexed: 11/17/2022] Open
Abstract
Intrauterine infection is considered as one of the major maternal insults during pregnancy. Intrauterine infection during pregnancy could lead to brain damage of the developmental fetus and offspring. Effects on the fetal, newborn, and adult central nervous system (CNS) may include signs of neurological problems, developmental abnormalities and delays, and intellectual deficits. However, the mechanisms or pathophysiology that leads to permanent brain damage during development are complex and not fully understood. This damage may affect morphogenic and behavioral phenotypes of the developed offspring, and that mice brain damage could be mediated through a final common pathway, which includes over-stimulation of excitatory amino acid receptor, over-production of vascularization/angiogenesis, pro-inflammatory cytokines, neurotrophic factors and apoptotic-inducing factors.
Collapse
Affiliation(s)
- Mahmoud Huleihel
- Department of Microbiology and Immunology and the BGU Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hava Golan
- Department of Microbiology and Immunology and the BGU Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Development and Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Mordechai Hallak
- Department of Microbiology and Immunology and the BGU Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Obstetrics & Gynecology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
40
|
Marín-Teva JL, Dusart I, Colin C, Gervais A, van Rooijen N, Mallat M. Microglia Promote the Death of Developing Purkinje Cells. Neuron 2004; 41:535-47. [PMID: 14980203 DOI: 10.1016/s0896-6273(04)00069-8] [Citation(s) in RCA: 562] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2003] [Revised: 10/13/2003] [Accepted: 01/20/2004] [Indexed: 11/17/2022]
Abstract
The loss of neuronal cells, a prominent event in the development of the nervous system, involves regulated triggering of programmed cell death, followed by efficient removal of cell corpses. Professional phagocytes, such as microglia, contribute to the elimination of dead cells. Here we provide evidence that, in addition to their phagocytic activity, microglia promote the death of developing neurons engaged in synaptogenesis. In the developing mouse cerebellum, Purkinje cells die, and 60% of these neurons that already expressed activated caspase-3 were engulfed or contacted by spreading processes emitted by microglial cells. Apoptosis of Purkinje cells in cerebellar slices was strongly reduced by selective elimination of microglia. Superoxide ions produced by microglial respiratory bursts played a major role in this Purkinje cell death. Our study illustrates a mammalian form of engulfment-promoted cell death that links the execution of neuron death to the scavenging of dead cells.
Collapse
Affiliation(s)
- José Luis Marín-Teva
- Biologie des Interactions Neurone-glie, INSERM U.495, IFR 70, UPMC, 47 Bd de l'hôpital, 75013 Paris, France
| | | | | | | | | | | |
Collapse
|
41
|
Hynds DL, Rangappa N, Ter Beest J, Snow DM, Rabchevsky AG. Microglia enhance dorsal root ganglion outgrowth in Schwann cell cultures. Glia 2004; 46:218-23. [PMID: 15042588 DOI: 10.1002/glia.10353] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Transplantation of cellular populations to facilitate regrowth of damaged axons is a common experimental therapy for spinal cord injury. Schwann cells (SC) or microglia grafted into injury sites can promote axonal regrowth of central projections of dorsal root ganglion (DRG) sensory neurons. We sought to determine whether the addition of microglia or microglia-derived secretory products alters DRG axon regrowth upon cultures of SC. Rat DRG explants were grown on monolayers consisting of either SC, microglia, SC exposed to microglia-conditioned medium (MCM), or co-cultures with different relative concentrations of microglia. Image analysis revealed that, compared to SC alone, the extent of neurite outgrowth was significantly greater on SC-microglia co-cultures. Immunocytochemistry for extracellular matrix molecules showed that microglial cells stained positively for growth-promoting thrombospondin, whereas laminin and the inhibitory chondroitin sulfate proteoglycans (CSPGs) were localized primarily to SC. Notably, immunoreactivity for CSPGs appeared reduced in areas associated with DRG outgrowth in co-cultures and SC exposed to MCM. These results show that microglia or their secreted products can augment SC-mediated DRG regrowth in vitro, indicating that co-grafting SC with microglia provides a novel approach to augment sensory fiber regeneration after spinal cord injury.
Collapse
Affiliation(s)
- Dianna L Hynds
- Department of Anatomy and Neurobiology, University of Kentucky Medical Center, Lexington, Kentucky 40536, USA
| | | | | | | | | |
Collapse
|
42
|
Rhodes KE, Moon LDF, Fawcett JW. Inhibiting cell proliferation during formation of the glial scar: effects on axon regeneration in the CNS. Neuroscience 2003; 120:41-56. [PMID: 12849739 DOI: 10.1016/s0306-4522(03)00285-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Following a CNS lesion many glial cell types proliferate and/or migrate to the lesion site, forming the glial scar. The majority of these cells express chondroitin sulphate proteoglycans (CS-PGs), previously shown to inhibit axonal growth. In this study, in an attempt to diminish glial scar formation and improve axonal regeneration, proliferating cells were eliminated from the lesion site. Adult rats received a continuous infusion of 2% cytosine-D-arabinofuranoside (araC) or saline for 7 days over the lesion site, immediately following a unilateral transection of the right medial forebrain bundle. Additional groups of rats that received subdural infusions prior to the lesion, and lesioned rats which received no infusion, were also compared in the analyses. Animals were killed at 4, 7, 12 or 18 days post-lesion (dpl) and immunohistochemistry was used to determine the effects of these treatments on tyrosine hydroxylase (TH)-lesioned axons, and on the injury response of glial cells. Almost complete elimination of NG2 oligodendrocyte progenitor cells from the lesion site was seen up to 7 dpl in araC-infused animals; reduced numbers of reactive CD11b microglia were also seen but no effects were seen on the injury response of GFAP astrocytes. Significantly more TH axons were seen distal to the lesion in araC-treated brains, but these numbers dwindled by 18 dpl.
Collapse
Affiliation(s)
- K E Rhodes
- Cambridge Centre for Brain Repair, University of Cambridge, E. D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 2PY, UK.
| | | | | |
Collapse
|
43
|
Luo Y, Cai J, Liu Y, Xue H, Chrest FJ, Wersto RP, Rao M. Microarray analysis of selected genes in neural stem and progenitor cells. J Neurochem 2002; 83:1481-97. [PMID: 12472902 DOI: 10.1046/j.1471-4159.2002.01260.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
To access and compare gene expression in fetal neuroepithelial cells (NEPs) and progenitor cells, we have used microarrays containing approximately 500 known genes related to cell cycle regulation, apoptosis, growth and differentiation. We have identified 152 genes that are expressed in NEPs and 209 genes expressed by progenitor cells. The majority of genes (141) detected in NEPs are also present in progenitor populations. There are 68 genes specifically expressed in progenitors with little or no expression in NEPs, and a few genes that appear to be present exclusively in NEPs. Using cell sorting, RT-PCR, in situ hybridization or immunocytochemistry, we have examined the segregation of expression to neuronal and glial progenitors, and identified several that appeared to be enriched in neuronal (e.g. CDK5, neuropilin, EphrinB2, FGF11) or glial (e.g. CXCR4, RhoC, CD44, tenascin C) precursors. Our data provide a first report of gene expression profiles of neural stem and progenitor cells at early stages of development, and provide evidence for the potential roles of specific cell cycle regulators, chemokines, cytokines and extracellular matrix molecules in neural development and lineage segregation.
Collapse
Affiliation(s)
- Yongquan Luo
- Laboratory of Neurosciences, Gerontology Research Center, National Institute on Aging, Room 4E02, 5600 Nathan Shock Drive, Baltimore, Maryland 21224, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Anlar B, Atilla P, Cakar AN, Kose MF, Beksaç MS, Dagdeviren A, Akçören Z. Expression of adhesion and extracellular matrix molecules in the developing human brain. J Child Neurol 2002; 17:707-13. [PMID: 12503652 DOI: 10.1177/088307380201700913] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cell adhesion molecules and extracellular matrix molecules have important roles in cell migration and connection. Their developmental expression has not been fully described in humans. In this report, these molecules were examined by immunohistochemistry in frontal tissue samples from 14- to 28-week-old fetuses aborted for obstetric reasons (n = 20) and four fetuses with nervous system abnormalities. Neural cell adhesion molecule (NCAM), tenascin, and laminin were expressed after 17 weeks. Neural cell adhesion molecule was observed in the neuropil, whereas tenascin and laminin also had cellular and vascular expression. Thrombospondin and fibronectin, apparent after 14 weeks, showed a redistribution from periventricular to outer cortical layers after midgestation. N-cadherin and integrin were observed in mid- and late gestation. Maternal or environmental conditions seemed to influence the pattern of expression. Fetuses with nervous system abnormalities had altered expression of several molecules. The descriptive data obtained in this study might constitute a basis for further studies investigating the role of these molecules in developmental abnormalities of the brain.
Collapse
Affiliation(s)
- Banu Anlar
- Department of Pediatric Neurology, Hacettepe University, Ankara, Turkey.
| | | | | | | | | | | | | |
Collapse
|
45
|
Rochefort N, Quenech'du N, Watroba L, Mallat M, Giaume C, Milleret C. Microglia and astrocytes may participate in the shaping of visual callosal projections during postnatal development. JOURNAL OF PHYSIOLOGY, PARIS 2002; 96:183-92. [PMID: 12445895 DOI: 10.1016/s0928-4257(02)00005-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the adult cat, axons running through the corpus callosum interconnect the border between the visual cortical areas 17 and 18 (A17 and A18) of both hemispheres. This specific pattern emerges during postnatal development, under normal viewing conditions (NR), from the elimination of initially exuberant callosal projections. In contrast, if the postnatal visual experience is monocular from birth (MD), juvenile callosal projections are stabilised throughout A17 and A18. The present study aimed at using such a model in vivo to find indications of a contribution of glial cells in the shaping of projections in the developing CNS through interactions with neurones, both in normal and pathological conditions. As a first stage, the distribution and the morphology of microglial cells and astrocytes were investigated from 2 weeks to adulthood. Microglial cells, stained with isolectin-B4, were clustered in the white matter below A17 and A18. Until one month, these clustered cells displayed an ameboid morphology in NR group, while they were more ramified in MD animals. Their phenotype thus depends on the postnatal visual experience, which indicates that microglial cells may interact with axons of visual neurones. It also suggests that they may differentially contribute to the elimination and the stabilisation of juvenile exuberant callosal fibres in NR and MD animals respectively. Beyond one month, microglial cells were very ramified in both experimental groups. Astrocytes were labelled with a GFAP-antibody. The distributions of connexins 43 (Cx43) and 30 (Cx30), the main proteic components of gap junction channels in astrocytes, were also investigated using specific antibodies. Both in NR and MD groups, until 1 month, GFAP-positive astrocytes and Cx43 were mainly localised within the subcortical white matter. Then GFAP, Cx43 and Cx30 stainings progressively appeared within the cortex, throughout A17 and A18 but with a differential laminar expression according to the age. Thus, the distributions of both astrocytes and connexins changed with age; however, the monocular occlusion had no visible effect. This suggests that astrocytes may contribute to the postnatal development of neuronal projections to the primary visual cortex, including visual callosal projections.
Collapse
Affiliation(s)
- N Rochefort
- Laboratoire de Physiologie de la Perception et de l'Action, UMR 7124, Collège de France, 11 Place Marcelin Berthelot, 75005 Paris, France
| | | | | | | | | | | |
Collapse
|
46
|
Dheen S, Hao A, Ng Y, Ling E. Regulatory Factors and Functions of Microglia during Development. Neuroembryology Aging 2002. [DOI: 10.1159/000063529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This article reviews current knowledge on the origin and development of microglia as well as on regulatory factors that influence microglial development within the neural tube during embryogenesis. Ameboid microglia found in the developing neural tube originate from mesodermal precursors derived from the yolk sac. These microglial cells, which exhibit characteristic features of reactive microglia, undergo mitosis in situ in the nervous parenchyma and function as the full-blown phagocytes involved in the removal of cellular debris resulting from neural tube defect or normal cellular turnover. During embryogenesis, the microglia express various cytokines, growth factors and chemokines. Some of those factors together with other local factors may influence the proliferation and activation of microglia in the developing neural tube.
Collapse
|
47
|
Abstract
An understanding of microglial functions during normal CNS development is prerequisite for understanding developmental neurotoxicology. This review provides a brief summary of previous work regarding the origin of microglia and addresses differences and similarities between microglia and brain macrophages. Current concepts and ideas which implicate microglia in diverse developmental processes, such as apoptosis, axon growth, and vasculogenesis are discussed. The study of reactive microgliosis may prove useful in the histopathological analysis of neurotoxicant-induced brain damage during development.
Collapse
Affiliation(s)
- W J Streit
- Department of Neuroscience, University of Florida College of Medicine and McKnight Brain Institute, Gainesville 32611, USA.
| |
Collapse
|
48
|
Cuadros MA, Navascués J. Early origin and colonization of the developing central nervous system by microglial precursors. PROGRESS IN BRAIN RESEARCH 2001; 132:51-9. [PMID: 11545016 DOI: 10.1016/s0079-6123(01)32065-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- M A Cuadros
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, E-18071 Granada, Spain.
| | | |
Collapse
|
49
|
Abstract
The postnatal development of rat microglia is marked by an important increase in the number of microglial cells and the growth of their ramified processes. We studied the role of thyroid hormone in microglial development. The distribution and morphology of microglial cells stained with isolectin B4 or monoclonal antibody ED1 were analyzed in cortical and subcortical forebrain regions of developing rats rendered hypothyroid by prenatal and postnatal treatment with methyl-thiouracil. Microglial processes were markedly less abundant in hypothyroid pups than in age-matched normal animals, from postnatal day 4 up to the end of the third postnatal week of life. A delay in process extension and a decrease in the density of microglial cell bodies, as shown by cell counts in the developing cingulate cortex of normal and hypothyroid animals, were responsible for these differences. Conversely, neonatal rat hyperthyroidism, induced by daily injections of 3,5,3'-triiodothyronine (T3), accelerated the extension of microglial processes and increased the density of cortical microglial cell bodies above physiological levels during the first postnatal week of life. Reverse transcription-PCR and immunological analyses indicated that cultured cortical ameboid microglial cells expressed the alpha1 and beta1 isoforms of nuclear thyroid hormone receptors. Consistent with the trophic and morphogenetic effects of thyroid hormone observed in situ, T3 favored the survival of cultured purified microglial cells and the growth of their processes. These results demonstrate that thyroid hormone promotes the growth and morphological differentiation of microglia during development.
Collapse
|
50
|
Vitkovic L, Maeda S, Sternberg E. Anti-inflammatory cytokines: expression and action in the brain. Neuroimmunomodulation 2001; 9:295-312. [PMID: 12045357 DOI: 10.1159/000059387] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Transforming growth factor-beta(1) (TGF-beta(1)) and interleukin (IL)-10 gene expression is equivocal in normal brain and upregulated in over a dozen central and peripheral diseases/disorders. The patterns of specific expression of cytokines differ in these diseases. Published data indicate that these cytokines are produced by and act on both neurons and glial cells. Although their actions are commonly viewed as 'anti-inflammatory', they protect neurons and downregulate the responses of glial cells to diseases/disorders in the absence of inflammation. Their actions counterbalance the actions of elevated IL-1 and/or tumor necrosis factor-alpha to maintain homeostasis. Their therapeutic potential will be realized by improving our understanding of their place in neural cytokine networks.
Collapse
Affiliation(s)
- L Vitkovic
- Integrative Neural Immune Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | | | | |
Collapse
|