101
|
Anderson SR, Vetter ML. Developmental roles of microglia: A window into mechanisms of disease. Dev Dyn 2019; 248:98-117. [PMID: 30444278 PMCID: PMC6328295 DOI: 10.1002/dvdy.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/21/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022] Open
Abstract
Microglia are engineers of the central nervous system (CNS) both in health and disease. In addition to the canonical immunological roles of clearing damaging entities and limiting the spread of toxicity and death, microglia remodel the CNS throughout life. While they have been extensively studied in disease and injury, due to their highly variable functions, their precise role in these contexts still remains uncertain. Over the past decade, we have greatly expanded our understanding of microglial function, including their essential homeostatic roles during development. Here, we review these developmental roles, identify parallels in disease, and speculate whether developmental mechanisms re-emerge in disease and injury. Developmental Dynamics 248:98-117, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sarah R Anderson
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, Utah
| | - Monica L Vetter
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
| |
Collapse
|
102
|
Webster KM, Sun M, Crack PJ, O'Brien TJ, Shultz SR, Semple BD. Age-dependent release of high-mobility group box protein-1 and cellular neuroinflammation after traumatic brain injury in mice. J Comp Neurol 2018; 527:1102-1117. [DOI: 10.1002/cne.24589] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/15/2018] [Accepted: 11/09/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Kyria M. Webster
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
| | - Mujun Sun
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
| | - Peter J. Crack
- Department of Pharmacology and Therapeutics; The University of Melbourne; Parkville Victoria Australia
| | - Terence J. O'Brien
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
- Department of Neuroscience; Monash University; Melbourne Victoria Australia
| | - Sandy R. Shultz
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
- Department of Neuroscience; Monash University; Melbourne Victoria Australia
| | - Bridgette D. Semple
- Department of Medicine (Royal Melbourne Hospital); The University of Melbourne; Parkville Victoria Australia
- Department of Neuroscience; Monash University; Melbourne Victoria Australia
| |
Collapse
|
103
|
Bergdolt L, Dunaevsky A. Brain changes in a maternal immune activation model of neurodevelopmental brain disorders. Prog Neurobiol 2018; 175:1-19. [PMID: 30590095 DOI: 10.1016/j.pneurobio.2018.12.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 12/13/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022]
Abstract
The developing brain is sensitive to a variety of insults. Epidemiological studies have identified prenatal exposure to infection as a risk factor for a range of neurological disorders, including autism spectrum disorder and schizophrenia. Animal models corroborate this association and have been used to probe the contribution of gene-environment interactions to the etiology of neurodevelopmental disorders. Here we review the behavior and brain phenotypes that have been characterized in MIA offspring, including the studies that have looked at the interaction between maternal immune activation and genetic risk factors for autism spectrum disorder or schizophrenia. These phenotypes include behaviors relevant to autism, schizophrenia, and other neurological disorders, alterations in brain anatomy, and structural and functional neuronal impairments. The link between maternal infection and these phenotypic changes is not fully understood, but there is increasing evidence that maternal immune activation induces prolonged immune alterations in the offspring's brain which could underlie epigenetic alterations which in turn may mediate the behavior and brain changes. These concepts will be discussed followed by a summary of the pharmacological interventions that have been tested in the maternal immune activation model.
Collapse
Affiliation(s)
- Lara Bergdolt
- University of Nebraska Medical Center, Neurological Sciences, 985960 Nebraska Medical Center, 68105, Omaha, NE, United States
| | - Anna Dunaevsky
- University of Nebraska Medical Center, Neurological Sciences, 985960 Nebraska Medical Center, 68105, Omaha, NE, United States.
| |
Collapse
|
104
|
Diz-Chaves Y, Toba L, Fandiño J, González-Matías LC, Garcia-Segura LM, Mallo F. The GLP-1 analog, liraglutide prevents the increase of proinflammatory mediators in the hippocampus of male rat pups submitted to maternal perinatal food restriction. J Neuroinflammation 2018; 15:337. [PMID: 30518432 PMCID: PMC6282252 DOI: 10.1186/s12974-018-1370-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/18/2018] [Indexed: 12/17/2022] Open
Abstract
Background Perinatal maternal malnutrition is related to altered growth of tissues and organs. The nervous system development is very sensitive to environmental insults, being the hippocampus a vulnerable structure, in which altered number of neurons and granular cells has been observed. Moreover, glial cells are also affected, and increased expression of proinflammatory mediators has been observed. We studied the effect of Glucagon-like peptide-1 receptor (GLP-1R) agonists, liraglutide, which have very potent metabolic and neuroprotective effects, in order to ameliorate/prevent the glial alterations present in the hippocampus of the pups from mothers with food restriction during pregnancy and lactation (maternal perinatal food restriction—MPFR). Methods Pregnant Sprague-Dawley rats were randomly assigned to 50% food restriction (FR; n = 12) or ad libitum controls (CT, n = 12) groups at day of pregnancy 12 (GD12). From GD14 to parturition, pregnant FR and CT rats were treated with liraglutide (100 μg/kg) or vehicle. At postnatal day 21 and before weaning, 48 males and 45 females (CT and MPFR) were sacrificed. mRNA expression levels of interleukin-1β (IL1β), interleukin-6 (IL-6), nuclear factor-κβ, major histocompatibility complex-II (MHCII), interleukin 10 (IL10), arginase 1 (Arg1), and transforming growth factor (TGFβ) were assessed in the hippocampus by quantitative real-time polymerase chain reaction. Iba1 and GFAP-immunoreactivity were assessed by immunocytochemistry. Results The mRNA expression IL1β, IL6, NF-κB, and MHCII increased in the hippocampus of male but not in female pups from MPFR. In addition, there was an increase in the percentage of GFAP and Iba1-immupositive cells in the dentate gyrus compared to controls, indicating an inflammatory response in the brain. On the other hand, liraglutide treatment prevented the neuroinflammatory process, promoting the production of anti-inflammatory molecules such as IL10, TGFβ, and arginase 1, and decreasing the number and reactivity of microglial cells and astrocytes in the hippocampus of male pups. Conclusion Therefore, the GLP-1 analog, liraglutide, emerges as neuroprotective drug that minimizes the harmful effects of maternal food restriction, decreasing neuroinflammation in the hippocampus in a very early stage.
Collapse
Affiliation(s)
- Y Diz-Chaves
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain.
| | - L Toba
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - J Fandiño
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - L C González-Matías
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, E-28002, Madrid, Spain.,Centro de Investigación en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - F Mallo
- Laboratory of Endocrinology, Biomedical Research Center (CINBIO), University of Vigo, Campus As Lagoas-Marcosende, E-36310, Vigo (Pontevedra), Spain
| |
Collapse
|
105
|
Luca A, Calandra C, Luca M. Molecular Bases of Alzheimer's Disease and Neurodegeneration: The Role of Neuroglia. Aging Dis 2018; 9:1134-1152. [PMID: 30574424 PMCID: PMC6284765 DOI: 10.14336/ad.2018.0201] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 02/01/2018] [Indexed: 12/13/2022] Open
Abstract
Neuroglia is an umbrella term indicating different cellular types that play a pivotal role in the brain, being involved in its development and functional homeostasis. Glial cells are becoming the focus of recent researches pertaining the pathogenesis of neurodegenerative disorders, Alzheimer's Disease (AD) in particular. In fact, activated microglia is the main determinant of neuroinflammation, contributing to neurodegeneration. In addition, the oxidative insult occurring during pathological brain aging can activate glial cells that, in turn, can favor the production of free radicals. Moreover, the recent Glycogen Synthase Kinase 3 (GSK-3) hypothesis of AD suggests that GSK3, involved in the regulation of glial cells functioning, could exert a role in amyloid deposition and tau hyper-phosphorylation. In this review, we briefly describe the main physiological functions of the glial cells and discuss the link between neuroglia and the most studied molecular bases of AD. In addition, we dedicate a section to the glial changes occurring in AD, with particular attention to their role in terms of neurodegeneration. In the light of the literature data, neuroglia could play a fundamental role in AD pathogenesis and progression. Further studies are needed to shed light on this topic.
Collapse
Affiliation(s)
- Antonina Luca
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University Hospital Policlinico-Vittorio Emanuele, Catania, 95100 Sicily, Italy
| | - Carmela Calandra
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University Hospital Policlinico-Vittorio Emanuele, Catania, 95100 Sicily, Italy
| | - Maria Luca
- Department of General Surgery and Medical-Surgical Specialties, Dermatology Clinic, University Hospital Policlinico-Vittorio Emanuele, Catania, 95100 Sicily, Italy
| |
Collapse
|
106
|
Edlow AG, Glass RM, Smith CJ, Tran PK, James K, Bilbo S. Placental Macrophages: A Window Into Fetal Microglial Function in Maternal Obesity. Int J Dev Neurosci 2018; 77:60-68. [PMID: 30465871 DOI: 10.1016/j.ijdevneu.2018.11.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/05/2018] [Accepted: 11/14/2018] [Indexed: 01/18/2023] Open
Abstract
Fetal placental macrophages and microglia (resident brain macrophages) have a common origin in the fetal yolk sac. Yolk-sac-derived macrophages comprise the permanent pool of brain microglia throughout an individual's lifetime. Inappropriate fetal microglial priming may therefore have lifelong neurodevelopmental consequences, but direct evaluation of microglial function in a living fetus or neonate is impossible. We sought to test the hypothesis that maternal obesity would prime both placental macrophages and fetal brain microglia to overrespond to an immune challenge, thus providing a window into microglial function using placental cells. Obesity was induced in C57BL/6 J mice using a 60% high-fat diet. On embryonic day 17.5, fetal brain microglia and corresponding CD11b + placental cells were isolated from fresh tissue. Cells were treated with media or lipopolysaccharide (LPS). Tumor necrosis factor-alpha (TNF-α) production by stimulated and unstimulated cells was quantified via ELISA. We demonstrate for the first time that the proinflammatory cytokine production of CD11b + placental cells is strongly correlated with that of brain microglia (Spearman's ρ = 0.73, p = 0.002) in the setting of maternal obesity. Maternal obesity-exposed CD11b + cells had an exaggerated response to LPS compared to controls, with a 5.1-fold increase in TNF-α production in placentas (p = 0.003) and 3.8-fold increase in TNF-α production in brains (p = 0.002). In sex-stratified analyses, only male obesity-exposed brains and placentas had significant increase in TNF-α production in response to LPS. Taken together, these data suggest that maternal obesity primes both placental macrophages and fetal brain microglia to overproduce a proinflammatory cytokine in response to immune challenge. Male brain and placental immune response is more marked than female in this setting. Given that fetal microglial priming may impact neuroimmune function throughout the lifespan, these data could provide insight into the male predominance of certain neurodevelopmental morbidities linked to maternal obesity, including cognitive dysfunction, autism spectrum disorder, and ADHD. Placental CD11b+ macrophages may have the potential to serve as an accessible biomarker of aberrant fetal brain immune activation in maternal obesity. This finding may have broader implications for assaying the impact of other maternal exposures on fetal brain development.
Collapse
Affiliation(s)
- Andrea G Edlow
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Massachusetts General Hospital, Vincent Center for Reproductive Biology
| | - Ruthy M Glass
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Massachusetts General Hospital, Vincent Center for Reproductive Biology
| | - Caroline J Smith
- Pediatrics and Program in Neuroscience, Harvard Medical School, Lurie Center for Autism, MassGeneral Hospital for Children
| | - Phuong Kim Tran
- Pediatrics and Program in Neuroscience, Harvard Medical School, Lurie Center for Autism, MassGeneral Hospital for Children
| | - Kaitlyn James
- Massachusetts General Hospital, Deborah Kelly Center for Outcomes Research
| | - Staci Bilbo
- Pediatrics and Program in Neuroscience, Harvard Medical School, Lurie Center for Autism, MassGeneral Hospital for Children
| |
Collapse
|
107
|
Labrousse VF, Leyrolle Q, Amadieu C, Aubert A, Sere A, Coutureau E, Grégoire S, Bretillon L, Pallet V, Gressens P, Joffre C, Nadjar A, Layé S. Dietary omega-3 deficiency exacerbates inflammation and reveals spatial memory deficits in mice exposed to lipopolysaccharide during gestation. Brain Behav Immun 2018; 73:427-440. [PMID: 29879442 DOI: 10.1016/j.bbi.2018.06.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/21/2018] [Accepted: 06/03/2018] [Indexed: 01/03/2023] Open
Abstract
Maternal immune activation (MIA) is a common environmental insult on the developing brain and represents a risk factor for neurodevelopmental disorders. Animal models of in utero inflammation further revealed a causal link between maternal inflammatory activation during pregnancy and behavioural impairment relevant to neurodevelopmental disorders in the offspring. Accumulating evidence point out that proinflammatory cytokines produced both in the maternal and fetal compartments are responsible for social, cognitive and emotional behavioral deficits in the offspring. Polyunsaturated fatty acids (PUFAs) are essential fatty acids with potent immunomodulatory activities. PUFAs and their bioactive derivatives can promote or inhibit many aspects of the immune and inflammatory response. PUFAs of the n-3 series ('n-3 PUFAs', also known as omega-3) exhibit anti-inflammatory/pro-resolution properties and promote immune functions, while PUFAs of the n-6 series ('n-6 PUFAs' or omega-6) favor pro-inflammatory responses. The present study aimed at providing insight into the effects of n-3 PUFAs on the consequences of MIA on brain development. We hypothesized that a reduction in n-3 PUFAs exacerbates both maternal and fetal inflammatory responses to MIA and later-life defects in memory in the offspring. Based on a lipopolysaccharide (LPS) model of MIA (LPS injection at embryonic day 17), we showed that n-3 PUFA deficiency 1) alters fatty acid composition of the fetal and adult offspring brain; 2) exacerbates maternal and fetal inflammatory processes with no significant alteration of microglia phenotype, and 3) induces spatial memory deficits in the adult offspring. We also showed a strong negative correlation between brain content in n-3 PUFA and cytokine production in MIA-exposed fetuses. Overall, our study is the first to address the deleterious effects of n-3 PUFA deficiency on brain lipid composition, inflammation and memory performances in MIA-exposed animals and indicates that it should be considered as a potent environmental risk factor for the apparition of neurodevelopmental disorders.
Collapse
Affiliation(s)
- V F Labrousse
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - Q Leyrolle
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France
| | - C Amadieu
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - A Aubert
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - A Sere
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - E Coutureau
- Centre National de la Recherche Scientifique, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Uité Mixte de Recherche 5287, 33076 Bordeaux, France; Université de Bordeaux, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, 33076 Bordeaux, France
| | - S Grégoire
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - L Bretillon
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - V Pallet
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - P Gressens
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - C Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - A Nadjar
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France.
| | - S Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France.
| |
Collapse
|
108
|
Cussotto S, Sandhu KV, Dinan TG, Cryan JF. The Neuroendocrinology of the Microbiota-Gut-Brain Axis: A Behavioural Perspective. Front Neuroendocrinol 2018; 51:80-101. [PMID: 29753796 DOI: 10.1016/j.yfrne.2018.04.002] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022]
Abstract
The human gut harbours trillions of symbiotic bacteria that play a key role in programming different aspects of host physiology in health and disease. These intestinal microbes are also key components of the gut-brain axis, the bidirectional communication pathway between the gut and the central nervous system (CNS). In addition, the CNS is closely interconnected with the endocrine system to regulate many physiological processes. An expanding body of evidence is supporting the notion that gut microbiota modifications and/or manipulations may also play a crucial role in the manifestation of specific behavioural responses regulated by neuroendocrine pathways. In this review, we will focus on how the intestinal microorganisms interact with elements of the host neuroendocrine system to modify behaviours relevant to stress, eating behaviour, sexual behaviour, social behaviour, cognition and addiction.
Collapse
Affiliation(s)
- Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Kiran V Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
109
|
Smolders S, Notter T, Smolders SMT, Rigo JM, Brône B. Controversies and prospects about microglia in maternal immune activation models for neurodevelopmental disorders. Brain Behav Immun 2018; 73:51-65. [PMID: 29870753 DOI: 10.1016/j.bbi.2018.06.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/26/2018] [Accepted: 06/01/2018] [Indexed: 12/16/2022] Open
Abstract
Activation of the maternal immune system during pregnancy is a well-established risk factor for neuropsychiatric disease in the offspring, yet, the underlying mechanisms leading to altered brain function remain largely undefined. Microglia, the resident immune cells of the brain, are key to adequate development of the central nervous system (CNS), and are prime candidates to mediate maternal immune activation (MIA)-induced brain abnormalities. As such, the effects of MIA on the immunological phenotype of microglia has been widely investigated. However, contradicting results due to differences in read-out and methodological approaches impede final conclusions on MIA-induced microglial alterations. The aim of this review is to critically discuss the evidence for an activated microglial phenotype upon MIA.
Collapse
Affiliation(s)
- Silke Smolders
- Uhasselt - BIOMED, Hasselt, Belgium; Laboratory of Neuronal Differentiation, VIB Center for the Biology of Disease, Leuven and Center for Human Genetics, KU Leuven Leuven, Belgium.
| | - Tina Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| | - Sophie M T Smolders
- Uhasselt - BIOMED, Hasselt, Belgium; INSERM, UMR S 1130, Université Pierre et Marie Curie Paris, France; CNRS, UMR 8246, Université Pierre et Marie Curie Paris, France; UM 119 NPS, Université Pierre et Marie Curie Paris, France.
| | | | | |
Collapse
|
110
|
Fernandez TV, Leckman JF, Pittenger C. Genetic susceptibility in obsessive-compulsive disorder. HANDBOOK OF CLINICAL NEUROLOGY 2018; 148:767-781. [PMID: 29478613 DOI: 10.1016/b978-0-444-64076-5.00049-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Obsessive-compulsive disorder (OCD) is present in 1.5-2.5% of the population and can result in substantial lifelong disability. It is characterized by intrusive thoughts, sensations, and urges and by repetitive behaviors that are difficult to control despite, in most cases, preserved insight as to their excessive or irrational nature. The causes and underlying pathophysiology of OCD are not well understood, which has limited the development of new treatments and interventions. Despite evidence for a substantial genetic contribution to disease risk, identification and replication of genetic variants associated with OCD have been challenging. Decades of candidate gene association studies have provided little insight. They are now being supplanted by modern genomewide approaches to discover both common and rare sequence and structural variants. Studies to date suggest potential novel therapeutic avenues such as modulators of glutamatergic and immune pathways; however, individual genetic findings are not yet statistically robust or replicated. Further efforts are clearly needed to identify specific risk variants and to confirm vulnerable pathways by studying much larger cohorts of patients with comprehensive variant discovery approaches. Mouse knockout models have already made notable inroads into our understanding of OCD pathology; their utility will only increase as specific risk alleles are identified.
Collapse
Affiliation(s)
- Thomas V Fernandez
- Child Study Center, Yale University School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States.
| | - James F Leckman
- Child Study Center, Yale University School of Medicine, New Haven, CT, United States; Department of Psychology, Yale University School of Medicine, New Haven, CT, United States
| | - Christopher Pittenger
- Child Study Center, Yale University School of Medicine, New Haven, CT, United States; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States; Department of Psychology, Yale University School of Medicine, New Haven, CT, United States; Integrated Neuroscience Research Program, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
111
|
Kagawa N, Nagao T. Neurodevelopmental toxicity in the mouse neocortex following prenatal exposure to acetamiprid. J Appl Toxicol 2018; 38:1521-1528. [PMID: 30047162 DOI: 10.1002/jat.3692] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/11/2018] [Accepted: 06/19/2018] [Indexed: 11/08/2022]
Abstract
Acetamiprid (ACE) belongs to a widely used class of pesticides known as neonicotinoids. ACE binds selectively to insect nicotinic acetylcholine receptors and was previously considered relatively safe in mammalian species; however, recent studies have demonstrated ACE-mediated toxicity related to vertebrate nicotinic acetylcholine receptor activation. The potential for neurotoxicity following exposure to ACE in utero is unknown. Therefore, we evaluated the effects of repeated prenatal ACE exposure (5 mg/kg, oral doses administered to pregnant dams) on neurogenesis, neuronal distribution and microglial activation in the dorsal telencephalon on embryonic day (E)14 and in the neocortex on postnatal day 14. Immunohistochemical and morphological analyses on E14 revealed hypoplasia of the cortical plate and decreased neurogenesis in mice exposed to ACE from E6 to E13, whereas newborn ACE-exposed mice showed an abnormal neuronal distribution in the neocortex. Additionally, ACE-exposed mice showed increased numbers of Iba1-immunoreactive and amoeboid-type microglia as well as an increased M1/M2 microglial ratio. These findings suggest that prenatal ACE exposure induces neurodevelopmental toxicity and increases microglial activation in the developing brain.
Collapse
Affiliation(s)
- Nao Kagawa
- Laboratory of Developmental Biology, Department of Life Sciences, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan
| | - Tetsuji Nagao
- Laboratory of Developmental Biology, Department of Life Sciences, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan
| |
Collapse
|
112
|
Microglia Are Critical in Host Defense against Prion Disease. J Virol 2018; 92:JVI.00549-18. [PMID: 29769333 DOI: 10.1128/jvi.00549-18] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/07/2018] [Indexed: 12/21/2022] Open
Abstract
Microglial cells in the central nervous system play important roles in neurodevelopment and resistance to infection, yet microglia can become neurotoxic under some conditions. An early event during prion infection is the activation of microglia and astrocytes in the brain prior to damage or death of neurons. Previous prion disease studies using two different strategies to manipulate signaling through the microglial receptor CSF-1R reported contrary effects on survival from prion disease. However, in these studies, reductions of microglial numbers and function were variable, thus confounding interpretation of the results. In the present work, we used oral treatment with a potent inhibitor of CSF-1R, PLX5622, to eliminate 78 to 90% of microglia from cortex early during the course of prion infection. Oral drug treatment early after infection with the RML scrapie strain significantly accelerated vacuolation, astrogliosis, and deposition of disease-associated prion protein. Furthermore, drug-treated mice had advanced clinical disease requiring euthanasia 31 days earlier than untreated control mice. Similarly, PLX5622 treatment during the preclinical phase at 80 days postinfection with RML scrapie also accelerated disease and resulted in euthanasia of mice 33 days earlier than infected controls. PLX5622 also accelerated clinical disease after infection with scrapie strains ME7 and 22L. Thus, microglia are critical in host defense during prion disease. The early accumulation of PrPSc in the absence of microglia suggested that microglia may function by clearing PrPSc, resulting in longer survival.IMPORTANCE Microglia contribute to many aspects of health and disease. When activated, microglia can be beneficial by repairing damage in the central nervous system (CNS) or they can turn harmful by becoming neurotoxic. In prion and prionlike diseases, the involvement of microglia in disease is unclear. Previous studies suggest that microglia can either speed up or slow down disease. In this study, we infected mice with prions and depleted microglia from the brains of mice using PLX5622, an effective CSF-1R tyrosine kinase inhibitor. Microglia were markedly reduced in brains, and prion disease was accelerated, so that mice needed to be euthanized 20 to 33 days earlier than infected control mice due to advanced clinical disease. Similar results occurred when mice were treated with PLX5622 at 80 days after infection, which was just prior to the start of clinical signs. Thus, microglia are important for removing prions, and the disease is faster when microglia are depleted.
Collapse
|
113
|
Qin Y, Garrison BS, Ma W, Wang R, Jiang A, Li J, Mistry M, Bronson RT, Santoro D, Franco C, Robinton DA, Stevens B, Rossi DJ, Lu C, Springer TA. A Milieu Molecule for TGF-β Required for Microglia Function in the Nervous System. Cell 2018; 174:156-171.e16. [PMID: 29909984 DOI: 10.1016/j.cell.2018.05.027] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/28/2018] [Accepted: 05/11/2018] [Indexed: 01/20/2023]
Abstract
Extracellular proTGF-β is covalently linked to "milieu" molecules in the matrix or on cell surfaces and is latent until TGF-β is released by integrins. Here, we show that LRRC33 on the surface of microglia functions as a milieu molecule and enables highly localized, integrin-αVβ8-dependent TGF-β activation. Lrrc33-/- mice lack CNS vascular abnormalities associated with deficiency in TGF-β-activating integrins but have microglia with a reactive phenotype and after 2 months develop ascending paraparesis with loss of myelinated axons and death by 5 months. Whole bone marrow transplantation results in selective repopulation of Lrrc33-/- brains with WT microglia and halts disease progression. The phenotypes of WT and Lrrc33-/- microglia in the same brain suggest that there is little spreading of TGF-β activated from one microglial cell to neighboring microglia. Our results suggest that interactions between integrin-bearing cells and cells bearing milieu molecule-associated TGF-β provide localized and selective activation of TGF-β.
Collapse
Affiliation(s)
- Yan Qin
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Brian S Garrison
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Department of Stem Cell and Regenerative Biology, Boston, MA 02115, USA
| | - Wenjiang Ma
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Rui Wang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Aiping Jiang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Meeta Mistry
- Harvard School of Public Health, Boston, MA 02115, USA
| | | | - Daria Santoro
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Charlotte Franco
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Daisy A Robinton
- Harvard Medical School, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Beth Stevens
- Harvard Medical School, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Derrick J Rossi
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Department of Stem Cell and Regenerative Biology, Boston, MA 02115, USA
| | - Chafen Lu
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
114
|
New Insights into Microglia-Neuron Interactions: A Neuron's Perspective. Neuroscience 2018; 405:103-117. [PMID: 29753862 DOI: 10.1016/j.neuroscience.2018.04.046] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/26/2018] [Accepted: 04/28/2018] [Indexed: 01/16/2023]
Abstract
Microglia are the primary immune cells of the central nervous system. However, recent data indicate that microglia also contribute to diverse physiological and pathophysiological processes that extend beyond immune-related functions and there is a growing interest to understand the mechanisms through which microglia interact with other cells in the brain. In particular, the molecular processes that contribute to microglia-neuron communication in the healthy brain and their role in common brain diseases have been intensively studied during the last decade. In line with this, fate-mapping studies, genetic models and novel pharmacological approaches have revealed the origin of microglial progenitors, demonstrated the role of self-maintaining microglial populations during brain development or in adulthood, and identified the unexpectedly long lifespan of microglia that may profoundly change our view about senescence and age-related human diseases. Despite the exponentially increasing knowledge about microglia, the role of these cells in health and disease is still extremely controversial and the precise molecular targets for intervention are not well defined. This is in part due to the lack of microglia-specific manipulation approaches until very recently and to the high level of complexity of the interactions between microglia and other cells in the brain that occur at different temporal and spatial scales. In this review, we briefly summarize the known physiological roles of microglia-neuron interactions in brain homeostasis and attempt to outline some major directions and challenges of future microglia research.
Collapse
|
115
|
Studying the Brain in a Dish: 3D Cell Culture Models of Human Brain Development and Disease. Curr Top Dev Biol 2018; 129:99-122. [PMID: 29801532 DOI: 10.1016/bs.ctdb.2018.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The study of the cellular and molecular processes of the developing human brain has been hindered by access to suitable models of living human brain tissue. Recently developed 3D cell culture models offer the promise of studying fundamental brain processes in the context of human genetic background and species-specific developmental mechanisms. Here, we review the current state of 3D human brain organoid models and consider their potential to enable investigation of complex aspects of human brain development and the underpinning of human neurological disease.
Collapse
|
116
|
|
117
|
Rübsam A, Parikh S, Fort PE. Role of Inflammation in Diabetic Retinopathy. Int J Mol Sci 2018; 19:ijms19040942. [PMID: 29565290 PMCID: PMC5979417 DOI: 10.3390/ijms19040942] [Citation(s) in RCA: 495] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/09/2018] [Accepted: 03/17/2018] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy is a common complication of diabetes and remains the leading cause of blindness among the working-age population. For decades, diabetic retinopathy was considered only a microvascular complication, but the retinal microvasculature is intimately associated with and governed by neurons and glia, which are affected even prior to clinically detectable vascular lesions. While progress has been made to improve the vascular alterations, there is still no treatment to counteract the early neuro-glial perturbations in diabetic retinopathy. Diabetes is a complex metabolic disorder, characterized by chronic hyperglycemia along with dyslipidemia, hypoinsulinemia and hypertension. Increasing evidence points to inflammation as one key player in diabetes-associated retinal perturbations, however, the exact underlying molecular mechanisms are not yet fully understood. Interlinked molecular pathways, such as oxidative stress, formation of advanced glycation end-products and increased expression of vascular endothelial growth factor have received a lot of attention as they all contribute to the inflammatory response. In the current review, we focus on the involvement of inflammation in the pathophysiology of diabetic retinopathy with special emphasis on the functional relationships between glial cells and neurons. Finally, we summarize recent advances using novel targets to inhibit inflammation in diabetic retinopathy.
Collapse
Affiliation(s)
- Anne Rübsam
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA.
| | - Sonia Parikh
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA.
| | - Patrice E Fort
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
118
|
Hilu-Dadia R, Hakim-Mishnaevski K, Levy-Adam F, Kurant E. Draper-mediated JNK signaling is required for glial phagocytosis of apoptotic neurons during Drosophila metamorphosis. Glia 2018. [PMID: 29520845 DOI: 10.1002/glia.23322] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Development of the central nervous system involves elimination of superfluous neurons through apoptosis and subsequent phagocytosis. In Drosophila, this occurs mainly during three developmental stages: embryogenesis, metamorphosis and emerging adult. Two transmembrane glial phagocytic receptors, SIMU (homolog of the mammalian Stabilin-2) and Draper (homolog of the mammalian MEGF10 and Jedi), mediate glial phagocytosis of apoptotic neurons during embryogenesis. However, less is known about the removal of apoptotic neurons during later stages of development. Here we show that during metamorphosis, Draper plays a critical role in apoptotic cell clearance by glia, whereas SIMU, which is mostly expressed in pupal macrophages outside the brain, is not involved in glial phagocytosis. We found that Draper activates Drosophila c-Jun N-terminal kinase (dJNK) signaling predominantly in the ensheathing glia and astrocytes, where it is required for efficient removal of apoptotic neurons. Our data suggest that besides the dJNK pathway, Draper also triggers an additional signaling pathway capable of removing apoptotic neurons in the pupal brain. This study thus reveals that SIMU unexpectedly is not involved in glial phagocytosis of apoptotic neurons during metamorphosis and highlights the novel role of dJNK signaling in developmental apoptotic cell clearance downstream of Draper.
Collapse
Affiliation(s)
- Reut Hilu-Dadia
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, 34988, Israel.,Department of Genetics and Developmental Biology, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, 31096, Israel
| | - Ketty Hakim-Mishnaevski
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, 34988, Israel
| | - Flonia Levy-Adam
- Department of Genetics and Developmental Biology, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, 31096, Israel
| | - Estee Kurant
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, 34988, Israel.,Department of Genetics and Developmental Biology, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, 31096, Israel
| |
Collapse
|
119
|
Bisphenol A exposure induces increased microglia and microglial related factors in the murine embryonic dorsal telencephalon and hypothalamus. Toxicol Lett 2018; 284:113-119. [DOI: 10.1016/j.toxlet.2017.12.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/24/2017] [Accepted: 12/13/2017] [Indexed: 11/22/2022]
|
120
|
Derk J, MacLean M, Juranek J, Schmidt AM. The Receptor for Advanced Glycation Endproducts (RAGE) and Mediation of Inflammatory Neurodegeneration. JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2018; 8:421. [PMID: 30560011 PMCID: PMC6293973 DOI: 10.4172/2161-0460.1000421] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Julia Derk
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, 550 First Avenue, Smilow 906, New York, NY, 10016, USA
| | - Michael MacLean
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, 550 First Avenue, Smilow 906, New York, NY, 10016, USA
| | - Judyta Juranek
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, 550 First Avenue, Smilow 906, New York, NY, 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, 550 First Avenue, Smilow 906, New York, NY, 10016, USA
| |
Collapse
|
121
|
Abstract
Microglia are the resident macrophages of the central nervous system parenchyma and fulfill crucial roles in brain development, homeostasis, and inflammation. The isolation of a pure microglia population from brain tissue enables the examination of microglial phenotypes without the interference of other cell populations. Microglial extractions from the neonatal brain have been described in various protocols, yet the more established and complex adult mouse brain poses a greater challenge. Here we describe a refined protocol including enzymatic and mechanical dissociation of adult mouse brain tissue and removal of myelin by Percoll density gradient. Microglial cells were subsequently extracted by an immunomagnetic approach. This isolation procedure enables the use of functionally viable cells for various applications such as cell culture, flow cytometry, functional assays including bacteria- or bead-based phagocytosis, stimulation assays, and transcriptome profiling techniques such as qRT-PCR and microarray/RNA sequencing.
Collapse
Affiliation(s)
- Kathleen Grabert
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, UK.
| | - Barry W McColl
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, UK
- Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
122
|
Abstract
The microbial ecosystem that inhabits the gastrointestinal tract of all mammals-the gut microbiota-has been in a symbiotic relationship with its hosts over many millennia. Thanks to modern technology, the myriad of functions that are controlled or modulated by the gut microbiota are beginning to unfold. One of the systems that is emerging to closely interact with the gut microbiota is the body's major neuroendocrine system that controls various body processes in response to stress, the hypothalamic-pituitary-adrenal (HPA) axis. This interaction is of pivotal importance; as various disorders of the microbiota-gut-brain axis are associated with dysregulation of the HPA axis. The present contribution describes the bidirectional communication between the gut microbiota and the HPA axis and delineates the potential underlying mechanisms. In this regard, it is important to note that the communication between the gut microbiota and the HPA axis is closely interrelated with other systems, such as the immune system, the intestinal barrier and blood-brain barrier, microbial metabolites, and gut hormones, as well as the sensory and autonomic nervous systems. These communication pathways will be exemplified through preclinical models of early life stress, beneficial roles of probiotics and prebiotics, evidence from germ-free mice, and antibiotic-induced modulation of the gut microbiota.
Collapse
Affiliation(s)
- Aitak Farzi
- Otto Loewi Research Center, Pharmacology Section, Medical University of Graz, Graz, Austria.
| | - Esther E Fröhlich
- Otto Loewi Research Center, Pharmacology Section, Medical University of Graz, Graz, Austria
| | - Peter Holzer
- Otto Loewi Research Center, Pharmacology Section, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
123
|
The Role of Microglia in Diabetic Retinopathy: Inflammation, Microvasculature Defects and Neurodegeneration. Int J Mol Sci 2018; 19:ijms19010110. [PMID: 29301251 PMCID: PMC5796059 DOI: 10.3390/ijms19010110] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 12/23/2017] [Accepted: 12/25/2017] [Indexed: 12/15/2022] Open
Abstract
Diabetic retinopathy is a common complication of diabetes mellitus, which appears in one third of all diabetic patients and is a prominent cause of vision loss. First discovered as a microvascular disease, intensive research in the field identified inflammation and neurodegeneration to be part of diabetic retinopathy. Microglia, the resident monocytes of the retina, are activated due to a complex interplay between the different cell types of the retina and diverse pathological pathways. The trigger for developing diabetic retinopathy is diabetes-induced hyperglycemia, accompanied by leukostasis and vascular leakages. Transcriptional changes in activated microglia, mediated via the nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) and extracellular signal–regulated kinase (ERK) signaling pathways, results in release of various pro-inflammatory mediators, including cytokines, chemokines, caspases and glutamate. Activated microglia additionally increased proliferation and migration. Among other consequences, these changes in microglia severely affected retinal neurons, causing increased apoptosis and subsequent thinning of the nerve fiber layer, resulting in visual loss. New potential therapeutics need to interfere with these diabetic complications even before changes in the retina are diagnosed, to prevent neuronal apoptosis and blindness in patients.
Collapse
|
124
|
Radtke FA, Chapman G, Hall J, Syed YA. Modulating Neuroinflammation to Treat Neuropsychiatric Disorders. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5071786. [PMID: 29181395 PMCID: PMC5664241 DOI: 10.1155/2017/5071786] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/13/2017] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is recognised as one of the potential mechanisms mediating the onset of a broad range of psychiatric disorders and may contribute to nonresponsiveness to current therapies. Both preclinical and clinical studies have indicated that aberrant inflammatory responses can result in altered behavioral responses and cognitive deficits. In this review, we discuss the role of inflammation in the pathogenesis of neuropsychiatric disorders and ask the question if certain genetic copy-number variants (CNVs) associated with psychiatric disorders might play a role in modulating inflammation. Furthermore, we detail some of the potential treatment strategies for psychiatric disorders that may operate by altering inflammatory responses.
Collapse
Affiliation(s)
- Franziska A. Radtke
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Gareth Chapman
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Yasir A. Syed
- Neuroscience and Mental Health Research Institute and School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| |
Collapse
|
125
|
Victorino AB, Serra FT, Piñero PP, de Almeida AA, Lopim GM, Matias Junior I, Machado HR, Lent R, Cabral FR, Gomez-Pinilla F, Arida RM, Gomes da Silva S. Aerobic exercise in adolescence results in an increase of neuronal and non-neuronal cells and in mTOR overexpression in the cerebral cortex of rats. Neuroscience 2017; 361:108-115. [PMID: 28802917 DOI: 10.1016/j.neuroscience.2017.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 07/29/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022]
Abstract
Better cognitive performance and greater cortical and hippocampal volume have been observed in individuals who undertook aerobic exercise during childhood and adolescence. One possible explanation for these beneficial effects is that juvenile physical exercise enables better neural development and hence more cells and neuronal circuitries. It is probable that such effects occur through intracellular signaling proteins associated with cell growth, proliferation and survival. Based on this information, we evaluated the number of neuronal and non-neuronal cells using isotropic fractionation and the expression and activation of intracellular proteins (ERK, CREB, Akt, mTOR and p70S6K) in the cerebral cortex and hippocampal formation of the rats submitted to a physical exercise program on a treadmill during adolescence. Results showed that physical exercise increases the number of neuronal and non-neuronal cortical cells and hippocampal neuronal cells in adolescent rats. Moreover, mTOR overexpression was found in the cortical region of exercised adolescent rats. These findings indicate a significant cellular proliferative effect of aerobic exercise on the cerebral cortex in postnatal development.
Collapse
Affiliation(s)
| | | | | | - Alexandre Aparecido de Almeida
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil; Instituto Federal Goiano (IF Goiano), Campus Ceres, Ceres, GO, Brazil
| | | | - Ivair Matias Junior
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), São Paulo, SP, Brazil
| | - Helio Rubens Machado
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), São Paulo, SP, Brazil
| | - Roberto Lent
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | | | | | | | - Sérgio Gomes da Silva
- Universidade de Mogi das Cruzes (UMC), Mogi das Cruzes, SP, Brazil; Hospital Israelita Albert Einstein (HIAE), São Paulo, SP, Brazil.
| |
Collapse
|
126
|
Decreased NOX2 expression in the brain of patients with bipolar disorder: association with valproic acid prescription and substance abuse. Transl Psychiatry 2017; 7:e1206. [PMID: 28809856 PMCID: PMC5611741 DOI: 10.1038/tp.2017.175] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/05/2017] [Accepted: 06/13/2017] [Indexed: 12/19/2022] Open
Abstract
Neuroinflammation and increased oxidative stress are believed to contribute to the development of psychiatric diseases. Animal studies have implicated NADPH oxidases (NOX) as relevant sources of reactive oxygen species in the brain. We have analyzed the expression of NOX isoforms in post-mortem brain samples from patients with psychiatric disorders (schizophrenia, bipolar disorder) and non-psychiatric subjects. Two collections from the Stanley Medical Research Institute were studied: the Array Collection (RNA, 35 individuals per group), and a neuropathology consortium collection (paraffin-embedded sections, 15 individuals per group). Quantitative PCR analysis revealed expression of NOX2 and NOX4 in prefrontal cortex. No impact of psychiatric disease on NOX4 levels was detected. Remarkably, the expression of NOX2 was specifically decreased in prefrontal and cingulate cortices of bipolar patients, as compared with controls and schizophrenic patients. NOX2 expression was not statistically associated with demographic parameters and post-mortem interval, but correlated with brain pH. Immunostaining demonstrated that NOX2 was predominantly expressed in microglia, which was corroborated by a decrease in the microglial markers CD68 and CD11b in the cingulate cortex of bipolar disorder patients. The analysis of potentially confounding parameters showed association of valproic acid prescription and heavy substance abuse with lower levels of NOX2. Taken together, we did not observe changes of NOX2 in schizophrenic patients, but a marked decrease of microglial markers and NOX2 in the brain of bipolar patients. This might be an underlying feature of bipolar disorder and/or a consequence of valproic acid treatment and substance abuse.
Collapse
|
127
|
Komada M, Hara N, Kawachi S, Kawachi K, Kagawa N, Nagao T, Ikeda Y. Mechanisms underlying neuro-inflammation and neurodevelopmental toxicity in the mouse neocortex following prenatal exposure to ethanol. Sci Rep 2017; 7:4934. [PMID: 28694481 PMCID: PMC5504035 DOI: 10.1038/s41598-017-04289-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/11/2017] [Indexed: 12/31/2022] Open
Abstract
Fetal alcohol spectrum disorders (FASD) constitute a wide range of disorders that arise from prenatal exposure to ethanol (EtOH). However, detailed reports regarding the adverse effects of prenatal EtOH exposure on neocortical morphology and its underlying pathogenic mechanisms are limited. In the present study, we aimed to characterize the anatomical abnormalities of neocortical development and their correlation with microglial properties and neuro-inflammation in a mouse model of FASD. We evaluated the development and maturation of the neocortex in ICR mice prenatally exposed to 25% (w/v) EtOH using histological and molecular analyses. Reduced proliferation and excessive cell death were observed in the dorsal telencephalon. Abnormal neuronal distribution, layer formation, and dopaminergic neuronal projections were observed in the neocortex. Disruption of microglial differentiation (M1/M2 microglial ratio) and abnormal expression of pro-inflammatory and neurotrophic factors were induced, and these abnormalities were ameliorated by co-treatment with an anti-inflammatory drug (pioglitazone). FASD model mice displayed histological abnormalities, microglial abnormalities, and neuro-inflammation in both the embryonic and newborn stages. Thus, anti-inflammatory therapeutics may provide a novel preventive approach for the treatment of FASD.
Collapse
Affiliation(s)
- Munekazu Komada
- Department of Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi, 464-8650, Japan.
| | - Nao Hara
- Department of Life Science, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan
| | - Satoko Kawachi
- Department of Life Science, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan
| | - Kota Kawachi
- Department of Life Science, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan
| | - Nao Kagawa
- Department of Life Science, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan
| | - Tetsuji Nagao
- Department of Life Science, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan
| | - Yayoi Ikeda
- Department of Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi, 464-8650, Japan
| |
Collapse
|
128
|
Gnanasegaran N, Govindasamy V, Mani V, Abu Kasim NH. Neuroimmunomodulatory properties of DPSCs in anin vitromodel of Parkinson's disease. IUBMB Life 2017; 69:689-699. [DOI: 10.1002/iub.1655] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/16/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Nareshwaran Gnanasegaran
- Department of Restorative Dentistry; Faculty of Dentistry, University of Malaya; Kuala Lumpur Malaysia
| | - Vijayendran Govindasamy
- Department of Restorative Dentistry; Faculty of Dentistry, University of Malaya; Kuala Lumpur Malaysia
| | - Vasudevan Mani
- Department of Pharmacology and Toxicology; College of Pharmacy, Qassim University; Buraidah Kingdom of Saudi Arabia
| | - Noor Hayaty Abu Kasim
- Department of Restorative Dentistry; Faculty of Dentistry, University of Malaya; Kuala Lumpur Malaysia
| |
Collapse
|
129
|
Fernández de Cossío L, Guzmán A, van der Veldt S, Luheshi GN. Prenatal infection leads to ASD-like behavior and altered synaptic pruning in the mouse offspring. Brain Behav Immun 2017; 63:88-98. [PMID: 27697456 DOI: 10.1016/j.bbi.2016.09.028] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/23/2016] [Accepted: 09/29/2016] [Indexed: 11/18/2022] Open
Abstract
Environmental challenges to the maternal immune system during pregnancy have been associated with an increase in the frequency of neurodevelopmental disorders such as Autism Spectrum Disorders (ASD) appearing in the offspring. Microglia, the brain's resident immune-cells, are now known to be critically involved in normal brain development, shaping connections between neurons by pruning superfluous synaptic spines. Our aim was to investigate whether maternal infection during critical stages of gestation compromises the role of microglia in sculpting neuronal circuits. Using a mouse model of maternal immune activation (MIA) induced by bacterial Lipopolysaccharide (LPS), we assayed the offspring's behavior during postnatal development. Additionally, we quantified spines within the offspring's brain and assessed alterations in some molecular signals involved in pruning. LPS-induced MIA led to behavioral changes relevant to ASD in the offspring in the absence of gross neurological problems. Prenatal LPS resulted in a significant increase in the number of spines in the granule cells of the dentate gyrus, as well as a reduction in hippocampal expression of the fractalkine microglial receptor (CX3CR1), involved in mediating the pruning process in the offspring. Interestingly, these changes were only noted in the male progeny of the LPS challenged dams. These results provide an early indicator that microglial function is altered in the brain of offspring from immune challenged mothers and that the effects in the brain appear to be specific along sex lines.
Collapse
Affiliation(s)
| | - Andrea Guzmán
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Quebec, Canada
| | - Suzanne van der Veldt
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Quebec, Canada; Nutrition et Neurobiologie Intégrée, INRA UMR 1286, 33076 Bordeaux, France; University of Bordeaux, Bordeaux, France
| | - Giamal N Luheshi
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Quebec, Canada.
| |
Collapse
|
130
|
Abstract
Microglia are the principal resident immune cells in the central nervous system and are believed to be versatile players in both inflammatory and physiological contexts. On the one hand, in order to safeguard the microenvironment microglia can be rapidly activated by contact with microbial products or cell debris, thereby exerting the functions of innate immunity via phagocytosis and secretion of cytokines and chemokines. Conversely, microglia can also assist in brain development, synaptic plasticity and neural repair through the production of neurotrophic factors and clearance of myelin debris. It is now well accepted that the dysfunction of microglia and microglia-induced neuroinflammation are implicated in the occurrence and progression of many neurological diseases. Although the past decade has witnessed major progress in understanding of multi-tasking microglia, what remains largely enigmatic is the relative importance of microglia at different disease stages and how microglia should be targeted for optimal therapeutic efficacy. Notably, microglia depletion through genetic targeting or pharmacological therapies can be viewed as effective tools to stimulate new microglia to repopulate the central nervous system. Microglia depletion and subsequent repopulation at defined stages in various experimental animal model disorders allow us to extend our knowledge of molecular mechanisms, thus holding promise for designing strategies to resolve neuroinflammation and promote recovery. Herein we highlight the highly plastic and diverse phenotypes of microglia and outline the lessons learned from microglia depletion approaches.
Collapse
Affiliation(s)
- Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at Solna, CMM L8:04, Karolinska Sjukhuset, S-171 76, Stockholm, Sweden
| | - Robert A Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at Solna, CMM L8:04, Karolinska Sjukhuset, S-171 76, Stockholm, Sweden
| | - Xing-Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at Solna, CMM L8:04, Karolinska Sjukhuset, S-171 76, Stockholm, Sweden.
| |
Collapse
|
131
|
Abstract
The recent advances in cell-based therapies for the repair of the pigmented epithelium is providing additional impetus for the translation of photoreceptor transplantation to eventual clinical trials. The prospects for transplantation of photoreceptors as a potential therapy for the treatment of photoreceptor degeneration will depend on successfully addressing many critical issues in preclinical studies. Although most of the studies that have carried out transplants of photoreceptors have primarily used normal mice, there have been recent reports that have also shown some success following transplantation to mouse models of retinitis pigmentosa. However, while these results are promising, there are several key issues that require further investigation in order to better understand the optimum timing for transplantation, given the extensive remodeling of the retina that occurs in late stage disease.
Collapse
|
132
|
Microglial Interferon Signaling and White Matter. Neurochem Res 2017; 42:2625-2638. [PMID: 28540600 DOI: 10.1007/s11064-017-2307-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/14/2017] [Accepted: 05/18/2017] [Indexed: 01/17/2023]
Abstract
Microglia, the resident immune cells of the CNS, are primary regulators of the neuroimmune response to injury. Type I interferons (IFNs), including the IFNαs and IFNβ, are key cytokines in the innate immune system. Their activity is implicated in the regulation of microglial function both during development and in response to neuroinflammation, ischemia, and neurodegeneration. Data from numerous studies in multiple sclerosis (MS) and stroke suggest that type I IFNs can modulate the microglial phenotype, influence the overall neuroimmune milieu, regulate phagocytosis, and affect blood-brain barrier integrity. All of these IFN-induced effects result in numerous downstream consequences on white matter pathology and microglial reactivity. Dysregulation of IFN signaling in mouse models with genetic deficiency in ubiquitin specific protease 18 (USP18) leads to a severe neurological phenotype and neuropathological changes that include white matter microgliosis and pro-inflammatory gene expression in dystrophic microglia. A class of genetic disorders in humans, referred to as pseudo-TORCH syndrome (PTS) for the clinical resemblance to infection-induced TORCH syndrome, also show dysregulation of IFN signaling, which leads to severe neurological developmental disease. In these disorders, the excessive activation of IFN signaling during CNS development results in a destructive interferonopathy with similar induction of microglial dysfunction as seen in USP18 deficient mice. Other recent studies implicate "microgliopathies" more broadly in neurological disorders including Alzheimer's disease (AD) and MS, suggesting that microglia are a potential therapeutic target for disease prevention and/or treatment, with interferon signaling playing a key role in regulating the microglial phenotype.
Collapse
|
133
|
Gosselin D, Skola D, Coufal NG, Holtman IR, Schlachetzki JCM, Sajti E, Jaeger BN, O'Connor C, Fitzpatrick C, Pasillas MP, Pena M, Adair A, Gonda DD, Levy ML, Ransohoff RM, Gage FH, Glass CK. An environment-dependent transcriptional network specifies human microglia identity. Science 2017; 356:science.aal3222. [PMID: 28546318 DOI: 10.1126/science.aal3222] [Citation(s) in RCA: 809] [Impact Index Per Article: 101.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 05/12/2017] [Indexed: 12/12/2022]
Abstract
Microglia play essential roles in central nervous system (CNS) homeostasis and influence diverse aspects of neuronal function. However, the transcriptional mechanisms that specify human microglia phenotypes are largely unknown. We examined the transcriptomes and epigenetic landscapes of human microglia isolated from surgically resected brain tissue ex vivo and after transition to an in vitro environment. Transfer to a tissue culture environment resulted in rapid and extensive down-regulation of microglia-specific genes that were induced in primitive mouse macrophages after migration into the fetal brain. Substantial subsets of these genes exhibited altered expression in neurodegenerative and behavioral diseases and were associated with noncoding risk variants. These findings reveal an environment-dependent transcriptional network specifying microglia-specific programs of gene expression and facilitate efforts to understand the roles of microglia in human brain diseases.
Collapse
Affiliation(s)
- David Gosselin
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Dylan Skola
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Nicole G Coufal
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037-1002, USA.,Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Inge R Holtman
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA.,Department of Neuroscience, section Medical Physiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Eniko Sajti
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Baptiste N Jaeger
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037-1002, USA
| | - Carolyn O'Connor
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037-1002, USA
| | - Conor Fitzpatrick
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037-1002, USA
| | - Martina P Pasillas
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| | - Monique Pena
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037-1002, USA
| | - Amy Adair
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037-1002, USA
| | - David D Gonda
- Department of Neurosurgery, University of California, San Diego-Rady Children's Hospital, San Diego, CA 92123, USA
| | - Michael L Levy
- Department of Neurosurgery, University of California, San Diego-Rady Children's Hospital, San Diego, CA 92123, USA
| | | | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037-1002, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA. .,Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, USA
| |
Collapse
|
134
|
Ye J, Jiang Z, Chen X, Liu M, Li J, Liu N. The role of autophagy in pro-inflammatory responses of microglia activation via mitochondrial reactive oxygen species in vitro. J Neurochem 2017; 142:215-230. [PMID: 28407242 DOI: 10.1111/jnc.14042] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 03/26/2017] [Accepted: 03/31/2017] [Indexed: 12/11/2022]
Abstract
Microglia over-activation contributes to neurodegenerative processes by neurotoxin factors and pro-inflammatory molecules of pro-inflammatory processes. Mitochondrial reactive oxygen species (ROS) and autophagy pathway might be involved in microglial activation, but the underlying mechanism is unclear. Here, we regulated autophagy pathway of microglia in vitro by autophagy inhibition (3-methyladenine treatment, siRNA-Beclin 1 or siRNA-ATG5 transfection) or induction (rapamycin treatment) in murine microglial BV-2 cells or cultured primary mouse microglial cells. And we found that autophagy inhibition could sensitize mitochondrial profile and microglial activation of cultured microglial cells, demonstrated by significant production of mitochondrial ROS, loss of mitochondrial membrane potential, secretion of pro-inflammatory cytokines including interleukin 1β (IL-1β), interleukin 6 (IL-6), interleukin 12 (IL-12) and tumor necrosis factor α and marked activation of mitogen-activated proteinkinases (MAPKs) and nuclear factor κB (NF-κB). These effects could be blocked by specific inhibitors of MAPK and NF-κB or mitochondrial antioxidants, Mito-TEMPO. Meanwhile, induction of autophagy with rapamycin treatment could significantly suppress microglial inflammatory responses, mitochondrial ROS production, activation of MAPKs and NF-κB. Taken together, our in vitro results from primary cultured microglia and BV-2 cell lines indicated that autophagy inhibition might participate in brain macrophage or microglia over-activation and mitochondrial ROS generation might be involved in the regulatory microglial pro-inflammatory responses.
Collapse
Affiliation(s)
- Junli Ye
- Department of Pathophysiology, Medical College, Qingdao University, Qingdao, Shandong, China
| | - Zhongxin Jiang
- Department of Clinical Laboratory, the Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Xuehong Chen
- Department of Pharmacology, Medical College, Qingdao University, Qingdao, China
| | - Mengyang Liu
- Department of Clinical Laboratory, the Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Jing Li
- Department of Clinical Laboratory, the Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Na Liu
- Department of Clinical Laboratory, the Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| |
Collapse
|
135
|
Nomura K, Vilalta A, Allendorf DH, Hornik TC, Brown GC. Activated Microglia Desialylate and Phagocytose Cells via Neuraminidase, Galectin-3, and Mer Tyrosine Kinase. THE JOURNAL OF IMMUNOLOGY 2017; 198:4792-4801. [PMID: 28500071 PMCID: PMC5458330 DOI: 10.4049/jimmunol.1502532] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/12/2017] [Indexed: 12/31/2022]
Abstract
Activated microglia can phagocytose dying, stressed, or excess neurons and synapses via the phagocytic receptor Mer tyrosine kinase (MerTK). Galectin-3 (Gal-3) can cross-link surface glycoproteins by binding galactose residues that are normally hidden below terminal sialic acid residues. Gal-3 was recently reported to opsonize cells via activating MerTK. We found that LPS-activated BV-2 microglia rapidly released Gal-3, which was blocked by calcineurin inhibitors. Gal-3 bound to MerTK on microglia and to stressed PC12 (neuron-like) cells, and it increased microglial phagocytosis of PC12 cells or primary neurons, which was blocked by inhibition of MerTK. LPS-activated microglia exhibited a sialidase activity that desialylated PC12 cells and could be inhibited by Tamiflu, a neuraminidase (sialidase) inhibitor. Sialidase treatment of PC12 cells enabled Gal-3 to bind and opsonize the live cells for phagocytosis by microglia. LPS-induced microglial phagocytosis of PC12 was prevented by small interfering RNA knockdown of Gal-3 in microglia, lactose inhibition of Gal-3 binding, inhibition of neuraminidase with Tamiflu, or inhibition of MerTK by UNC569. LPS-induced phagocytosis of primary neurons by primary microglia was also blocked by inhibition of MerTK. We conclude that activated microglia release Gal-3 and a neuraminidase that desialylates microglial and PC12 surfaces, enabling Gal-3 binding to PC12 cells and their phagocytosis via MerTK. Thus, Gal-3 acts as an opsonin of desialylated surfaces, and inflammatory loss of neurons or synapses may potentially be blocked by inhibiting neuraminidases, Gal-3, or MerTK.
Collapse
Affiliation(s)
- Koji Nomura
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - Anna Vilalta
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - David H Allendorf
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - Tamara C Hornik
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| |
Collapse
|
136
|
Lee AS, Azmitia EC, Whitaker-Azmitia PM. Developmental microglial priming in postmortem autism spectrum disorder temporal cortex. Brain Behav Immun 2017; 62:193-202. [PMID: 28159644 DOI: 10.1016/j.bbi.2017.01.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/13/2017] [Accepted: 01/26/2017] [Indexed: 10/20/2022] Open
Abstract
Microglia can shift into different complex morphologies depending on the microenvironment of the central nervous system (CNS). The distinct morphologies correlate with specific functions and can indicate the pathophysiological state of the CNS. Previous postmortem studies of autism spectrum disorder (ASD) showed neuroinflammation in ASD indicated by increased microglial density. These changes in the microglia density can be accompanied by changes in microglia phenotype but the individual contribution of different microglia phenotypes to the pathophysiology of ASD remains unclear. Here, we used an unbiased stereological approach to quantify six structurally and functionally distinct microglia phenotypes in postmortem human temporal cortex, which were immuno-stained with Iba1. The total density of all microglia phenotypes did not differ between ASD donors and typically developing individual donors. However, there was a significant decrease in ramified microglia in both gray matter and white matter of ASD, and a significant increase in primed microglia in gray matter of ASD compared to typically developing individuals. This increase in primed microglia showed a positive correlation with donor age in both gray matter and white of ASD, but not in typically developing individuals. Our results provide evidence of a shift in microglial phenotype that may indicate impaired synaptic plasticity and a chronic vulnerability to exaggerated immune responses.
Collapse
Affiliation(s)
- Andrew S Lee
- Department of Psychology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Biology, New York University, New York, NY 10003, USA; Max Planck Institute for Biological Cybernetics, 72076 Tuebingen, Germany.
| | - Efrain C Azmitia
- Department of Biology, New York University, New York, NY 10003, USA
| | | |
Collapse
|
137
|
Gross A, Benninger F, Madar R, Illouz T, Griffioen K, Steiner I, Offen D, Okun E. Toll-like receptor 3 deficiency decreases epileptogenesis in a pilocarpine model of SE-induced epilepsy in mice. Epilepsia 2017; 58:586-596. [DOI: 10.1111/epi.13688] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Adi Gross
- The Mina and Everard Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat-Gan Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center; Bar-Ilan University; Ramat-Gan Israel
- The Paul Feder Laboratory on Alzheimer's disease research; Tel-Aviv University; Tel Aviv Israel
| | - Felix Benninger
- The Mina and Everard Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat-Gan Israel
- The Neuroscience Laboratory; Felsenstein Medical Research Center; Tel-Aviv University; Tel Aviv Israel
- Department of Neurology; Rabin Medical Center; Petach Tikva Israel
| | - Ravit Madar
- The Mina and Everard Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat-Gan Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center; Bar-Ilan University; Ramat-Gan Israel
- The Paul Feder Laboratory on Alzheimer's disease research; Tel-Aviv University; Tel Aviv Israel
| | - Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center; Bar-Ilan University; Ramat-Gan Israel
- The Paul Feder Laboratory on Alzheimer's disease research; Tel-Aviv University; Tel Aviv Israel
| | - Kathleen Griffioen
- Department of Biology and Chemistry; Liberty University; Lynchburg Virginia U.S.A
| | - Israel Steiner
- Department of Neurology; Rabin Medical Center; Petach Tikva Israel
| | - Daniel Offen
- The Neuroscience Laboratory; Felsenstein Medical Research Center; Tel-Aviv University; Tel Aviv Israel
| | - Eitan Okun
- The Mina and Everard Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat-Gan Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center; Bar-Ilan University; Ramat-Gan Israel
- The Paul Feder Laboratory on Alzheimer's disease research; Tel-Aviv University; Tel Aviv Israel
| |
Collapse
|
138
|
Horiuchi M, Smith L, Maezawa I, Jin LW. CX 3CR1 ablation ameliorates motor and respiratory dysfunctions and improves survival of a Rett syndrome mouse model. Brain Behav Immun 2017; 60:106-116. [PMID: 26883520 PMCID: PMC5531048 DOI: 10.1016/j.bbi.2016.02.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/09/2016] [Accepted: 02/13/2016] [Indexed: 01/22/2023] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by loss-of-function mutations in the gene encoding MeCP2, an epigenetic modulator that binds the methyl CpG dinucleotide in target genes to regulate transcription. Previously we and others reported a role of microglia in the pathophysiology of RTT. Because microglia in the Mecp2 knockout (Mecp2KO) mouse model of RTT over-produce neurotoxic mediators glutamate and reactive oxygen species, we hypothesize that blocking neuron-microglia interaction by ablation of CX3CR1, a chemokine receptor expressed in microglia/myeloid cells mediating such interaction by pairing with its neuronal ligand CX3CL1, would ameliorate the RTT-like phenotype in Mecp2KO mice. Here we report that CX3CR1 ablation prolonged the lifespan of Mecp2KO mice from a median survival of 54.5-74days, and significantly improved the body weight gain, symptomatic scores, major respiratory parameters, and motor coordination and performance. CX3CR1 ablation rectified previously identified histological abnormalities in the Mecp2KO brain such as neuronal soma size in hippocampal CA2, and the number, soma size, and process complexity of microglia. Moreover, CX3CR1 ablation enhanced the neurotrophic action of microglia in Mecp2KO mice by producing higher amount of insulin-like growth factor 1. Our data support a role of myeloid cells/microglia in RTT and suggest a novel therapeutic approach for RTT by targeting CX3CR1 with specific antagonists or genetic downregulation.
Collapse
Affiliation(s)
- Makoto Horiuchi
- Department of Pathology and Laboratory Medicine, 2805 50th Street, UC Davis Medical Center, Sacramento, CA 95817, United States
| | - Lucas Smith
- Department of Pathology and Laboratory Medicine, 2805 50th Street, UC Davis Medical Center, Sacramento, CA 95817, United States
| | - Izumi Maezawa
- Department of Pathology and Laboratory Medicine, 2805 50th Street, UC Davis Medical Center, Sacramento, CA 95817, United States,M.I.N.D. (Medical Investigation of Neurodevelopmental Disorders) Institute, 2805 50th Street, UC Davis Medical Center, Sacramento, CA 95817, United States
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, 2805 50th Street, UC Davis Medical Center, Sacramento, CA 95817, United States; M.I.N.D. (Medical Investigation of Neurodevelopmental Disorders) Institute, 2805 50th Street, UC Davis Medical Center, Sacramento, CA 95817, United States.
| |
Collapse
|
139
|
|
140
|
Pierre WC, Smith PLP, Londono I, Chemtob S, Mallard C, Lodygensky GA. Neonatal microglia: The cornerstone of brain fate. Brain Behav Immun 2017; 59:333-345. [PMID: 27596692 DOI: 10.1016/j.bbi.2016.08.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/30/2016] [Accepted: 08/29/2016] [Indexed: 12/16/2022] Open
Abstract
Microglia, mainly known for their role in innate immunity and modulation of neuroinflammation, play an active role in central nervous system development and homeostasis. Depending on the context and environmental stimuli, microglia adopt a broad spectrum of activation status from pro-inflammatory, associated with neurotoxicity, to anti-inflammatory linked to neuroprotection. Pro-inflammatory microglial activation is a key hallmark of white matter injury in preterm infants and is involved in developmental origin of adult neurological diseases. Characterization of neonatal microglia function in brain development and inflammation has allowed the investigation of promising therapeutic targets with potential long-lasting neuroprotective effects. True prevention of neuro-degenerative diseases might eventually occur as early as the perinatal period.
Collapse
Affiliation(s)
- Wyston C Pierre
- Sainte-Justine Hospital and Research Center, Department of Pediatrics, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada
| | - Peter L P Smith
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Irène Londono
- Sainte-Justine Hospital and Research Center, Department of Pediatrics, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada
| | - Sylvain Chemtob
- Sainte-Justine Hospital and Research Center, Department of Pediatrics, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada; Departments of Ophtalmology, Université de Montréal, Montreal, Quebec, Canada; Departments of Pharmacology, Université de Montréal, Montreal, Quebec, Canada
| | - Carina Mallard
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Gregory A Lodygensky
- Sainte-Justine Hospital and Research Center, Department of Pediatrics, Université de Montréal, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, Québec, Canada; Montreal Heart Institute, 5000 Rue Bélanger, Montreal, Quebec, Canada; Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada; Departments of Pharmacology, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
141
|
Kalsbeek MJT, Mulder L, Yi CX. Microglia energy metabolism in metabolic disorder. Mol Cell Endocrinol 2016; 438:27-35. [PMID: 27687525 DOI: 10.1016/j.mce.2016.09.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022]
Abstract
Microglia are the resident macrophages of the CNS, and are in charge of maintaining a healthy microenvironment to ensure neuronal survival. Microglia carry out a non-stop patrol of the CNS, make contact with neurons and look for abnormalities, all of which requires a vast amount of energy. This non-signaling energy demand increases after activation by pathogens, neuronal damage or other kinds of stimulation. Of the three major energy substrates - glucose, fatty acids and glutamine - glucose is crucial for microglia survival and several glucose transporters are expressed to supply sufficient glucose influx. Fatty acids are another source of energy for microglia and have also been shown to strongly influence microglial immune activity. Glutamine, although possibly suitable for use as an energy substrate by microglia, has been shown to have neurotoxic effects when overloaded. Microglial fuel metabolism might be associated with microglial reactivity under different pathophysiological conditions and a microglial fuel switch may thus be the underlying cause of hypothalamic dysregulation, which is associated with obesity.
Collapse
Affiliation(s)
- Martin J T Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam (UvA), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Laurie Mulder
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam (UvA), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam (UvA), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
142
|
Abstract
As the immune-competent cells of the brain, microglia play an increasingly important role in maintaining normal brain function. They invade the brain early in development, transform into a highly ramified phenotype, and constantly screen their environment. Microglia are activated by any type of pathologic event or change in brain homeostasis. This activation process is highly diverse and depends on the context and type of the stressor or pathology. Microglia can strongly influence the pathologic outcome or response to a stressor due to the release of a plethora of substances, including cytokines, chemokines, and growth factors. They are the professional phagocytes of the brain and help orchestrate the immunological response by interacting with infiltrating immune cells. We describe here the diversity of microglia phenotypes and their responses in health, aging, and disease. We also review the current literature about the impact of lifestyle on microglia responses and discuss treatment options that modulate microglial phenotypes.
Collapse
Affiliation(s)
- Susanne A Wolf
- Cellular Neurosciences, Max Delbrück Centre for Molecular Medicine in the Helmholtz Association, Berlin 13092, Germany;
| | - H W G M Boddeke
- Department of Neuroscience, University of Groningen, University Medical Center Groningen, Groningen 9713, The Netherlands
| | - Helmut Kettenmann
- Cellular Neurosciences, Max Delbrück Centre for Molecular Medicine in the Helmholtz Association, Berlin 13092, Germany;
| |
Collapse
|
143
|
Kwan V, Unda BK, Singh KK. Wnt signaling networks in autism spectrum disorder and intellectual disability. J Neurodev Disord 2016; 8:45. [PMID: 27980692 PMCID: PMC5137220 DOI: 10.1186/s11689-016-9176-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/07/2016] [Indexed: 12/20/2022] Open
Abstract
Background Genetic factors play a major role in the risk for neurodevelopmental disorders such as autism spectrum disorders (ASDs) and intellectual disability (ID). The underlying genetic factors have become better understood in recent years due to advancements in next generation sequencing. These studies have uncovered a vast number of genes that are impacted by different types of mutations (e.g., de novo, missense, truncation, copy number variations). Abstract Given the large volume of genetic data, analyzing each gene on its own is not a feasible approach and will take years to complete, let alone attempt to use the information to develop novel therapeutics. To make sense of independent genomic data, one approach is to determine whether multiple risk genes function in common signaling pathways that identify signaling “hubs” where risk genes converge. This approach has led to multiple pathways being implicated, such as synaptic signaling, chromatin remodeling, alternative splicing, and protein translation, among many others. In this review, we analyze recent and historical evidence indicating that multiple risk genes, including genes denoted as high-confidence and likely causal, are part of the Wingless (Wnt signaling) pathway. In the brain, Wnt signaling is an evolutionarily conserved pathway that plays an instrumental role in developing neural circuits and adult brain function. Conclusions We will also review evidence that pharmacological therapies and genetic mouse models further identify abnormal Wnt signaling, particularly at the synapse, as being disrupted in ASDs and contributing to disease pathology.
Collapse
Affiliation(s)
- Vickie Kwan
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8S 4K1 Canada
| | - Brianna K Unda
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8S 4K1 Canada
| | - Karun K Singh
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario L8S 4K1 Canada
| |
Collapse
|
144
|
Role of Microglia in Neurological Disorders and Their Potentials as a Therapeutic Target. Mol Neurobiol 2016; 54:7567-7584. [DOI: 10.1007/s12035-016-0245-0] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023]
|
145
|
The promises and challenges of human brain organoids as models of neuropsychiatric disease. Nat Med 2016; 22:1220-1228. [DOI: 10.1038/nm.4214] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/22/2016] [Indexed: 12/12/2022]
|
146
|
Schmidt AF, Kannan PS, Chougnet CA, Danzer SC, Miller LA, Jobe AH, Kallapur SG. Intra-amniotic LPS causes acute neuroinflammation in preterm rhesus macaques. J Neuroinflammation 2016; 13:238. [PMID: 27596440 PMCID: PMC5011884 DOI: 10.1186/s12974-016-0706-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/25/2016] [Indexed: 01/06/2023] Open
Abstract
Background Chorioamnionitis is associated with an increased risk of brain injury in preterm neonates. Inflammatory changes in brain could underlie this injury. Here, we evaluated whether neuroinflammation is induced by chorioamnionitis in a clinically relevant model. Methods Rhesus macaque fetuses were exposed to either intra-amniotic (IA) saline, or IA lipopolysaccharide (LPS) (1 mg) 16 or 48 h prior to delivery at 130 days (85 % of gestation) (n = 4–5 animals/group). We measured cytokines in the cerebrospinal fluid (CSF), froze samples from the left brain for molecular analysis, and immersion fixed the right brain hemisphere for immunohistology. We analyzed the messenger RNA (mRNA) levels of the pro-inflammatory cytokines IL-1β, CCL2, TNF-α, IL-6, IL-8, IL-10, and COX-2 in the periventricular white matter (PVWM), cortex, thalamus, hippocampus, and cerebellum by RT-qPCR. Brain injury was assessed by immunohistology for myelin basic protein (MBP), IBA1 (microglial marker), GFAP (astrocyte marker), OLIG2 (oligodendrocyte marker), NeuN (neuronal marker), CD3 (T cells), and CD14 (monocytes). Microglial proliferation was assessed by co-immunostaining for IBA1 and Ki67. Data were analyzed by ANOVA with Tukey’s post-test. Results IA LPS increased mRNA expression of pro-inflammatory cytokines in the PVWM, thalamus, and cerebellum, increased IL-6 concentration in the CSF, and increased apoptosis in the periventricular area after 16 h. Microglial proliferation in the white matter was increased 48 h after IA LPS. Conclusions LPS-induced chorioamnionitis caused neuroinflammation, microglial proliferation, and periventricular apoptosis in a clinically relevant model of chorioamnionitis in fetal rhesus macaques. These findings identify specific responses in the fetal brain and support the hypothesis that neuroinflammatory changes may mediate the adverse neurodevelopmental outcomes associated with chorioamnionitis. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0706-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Augusto F Schmidt
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Paranthaman S Kannan
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lisa A Miller
- California National Primate Research Center and Department of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - Alan H Jobe
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Suhas G Kallapur
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
| |
Collapse
|
147
|
Leckman JF, Fernandez TV. The Origins of Tourette Syndrome: Prenatal Risk Factors and the Promise of Birth Cohort Studies. J Am Acad Child Adolesc Psychiatry 2016; 55:751-3. [PMID: 27566115 PMCID: PMC5180440 DOI: 10.1016/j.jaac.2016.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/27/2016] [Indexed: 11/28/2022]
Affiliation(s)
- James F. Leckman
- Address correspondence to: James F. Leckman, MD, PhD,
230 S Frontage Road, New Haven, CT 06520, Tel: 203-785-5880, Fax: 203-737-5104,
| | | |
Collapse
|
148
|
Sorrentino FS, Allkabes M, Salsini G, Bonifazzi C, Perri P. The importance of glial cells in the homeostasis of the retinal microenvironment and their pivotal role in the course of diabetic retinopathy. Life Sci 2016; 162:54-9. [PMID: 27497914 DOI: 10.1016/j.lfs.2016.08.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 07/24/2016] [Accepted: 08/02/2016] [Indexed: 01/18/2023]
Abstract
Diabetic retinopathy (DR) is a remarkable microvascular complication of diabetes and it has been considered the leading cause of legal blindness in working-age adults in the world. Several overlapping and interrelated molecular pathways are involved in the development of this disease. DR is staged into different levels of severity, from the nonproliferative to the advanced proliferative form. Over the years the progression of DR evolves through a series of changes involving distinct types of specialized cells: neural, vascular and glial. Prior to the clinically observable vascular complications, hyperglycemia and inflammation affect retinal glial cells which undergo a wide range of structural and functional alterations. In this review, we provide an overview of the status of macroglia and microglia in the course of DR, trying to briefly take into account the complex biochemical mechanisms that affect the intimate relationship among neuroretina, vessels and glial cells.
Collapse
Affiliation(s)
| | - Michael Allkabes
- Department of Biomedical and Surgical Sciences, Division of Ophthalmology, University of Ferrara, Ferrara, Italy
| | - Giulia Salsini
- Department of Biomedical and Surgical Sciences, Division of Ophthalmology, University of Ferrara, Ferrara, Italy
| | - Claudio Bonifazzi
- Department of Biomedical and Surgical Sciences, Section of Human Physiology, University of Ferrara, Ferrara, Italy
| | - Paolo Perri
- Department of Biomedical and Surgical Sciences, Division of Ophthalmology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
149
|
Jiang X, Nardelli J. Cellular and molecular introduction to brain development. Neurobiol Dis 2016; 92:3-17. [PMID: 26184894 PMCID: PMC4720585 DOI: 10.1016/j.nbd.2015.07.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 07/07/2015] [Accepted: 07/09/2015] [Indexed: 12/13/2022] Open
Abstract
Advances in the study of brain development over the last decades, especially recent findings regarding the evolutionary expansion of the human neocortex, and large-scale analyses of the proteome/transcriptome in the human brain, have offered novel insights into the molecular mechanisms guiding neural maturation, and the pathophysiology of multiple forms of neurological disorders. As a preamble to reviews of this issue, we provide an overview of the cellular, molecular and genetic bases of brain development with an emphasis on the major mechanisms associated with landmarks of normal neural development in the embryonic stage and early postnatal life, including neural stem/progenitor cell proliferation, cortical neuronal migration, evolution and folding of the cerebral cortex, synaptogenesis and neural circuit development, gliogenesis and myelination. We will only briefly depict developmental disorders that result from perturbations of these cellular or molecular mechanisms, and the most common perinatal brain injuries that could disturb normal brain development.
Collapse
Affiliation(s)
- Xiangning Jiang
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA
| | - Jeannette Nardelli
- Inserm, U1141, Paris 75019, France; Université Paris Diderot, Sorbonne Paris Cité, UMRS 1141, Paris 75019, France.
| |
Collapse
|
150
|
Milior G, Lecours C, Samson L, Bisht K, Poggini S, Pagani F, Deflorio C, Lauro C, Alboni S, Limatola C, Branchi I, Tremblay ME, Maggi L. Fractalkine receptor deficiency impairs microglial and neuronal responsiveness to chronic stress. Brain Behav Immun 2016; 55:114-125. [PMID: 26231972 DOI: 10.1016/j.bbi.2015.07.024] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/25/2015] [Accepted: 07/26/2015] [Indexed: 12/25/2022] Open
Abstract
Chronic stress is one of the most relevant triggering factors for major depression. Microglial cells are highly sensitive to stress and, more generally, to environmental challenges. However, the role of these brain immune cells in mediating the effects of stress is still unclear. Fractalkine signaling - which comprises the chemokine CX3CL1, mainly expressed by neurons, and its receptor CX3CR1, almost exclusively present on microglia in the healthy brain - has been reported to critically regulate microglial activity. Here, we investigated whether interfering with microglial function by deleting the Cx3cr1 gene affects the brain's response to chronic stress. To this purpose, we housed Cx3cr1 knockout and wild-type adult mice in either control or stressful environments for 2weeks, and investigated the consequences on microglial phenotype and interactions with synapses, synaptic transmission, behavioral response and corticosterone levels. Our results show that hampering neuron-microglia communication via the CX3CR1-CX3CL1 pathway prevents the effects of chronic unpredictable stress on microglial function, short- and long-term neuronal plasticity and depressive-like behavior. Overall, the present findings suggest that microglia-regulated mechanisms may underlie the differential susceptibility to stress and consequently the vulnerability to diseases triggered by the experience of stressful events, such as major depression.
Collapse
Affiliation(s)
- Giampaolo Milior
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| | - Cynthia Lecours
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada
| | - Louis Samson
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada
| | - Kanchan Bisht
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada
| | - Silvia Poggini
- Section of Behavioural Neurosciences, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Pagani
- Center for Life Nanoscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | - Cristina Deflorio
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy; Département de Neuroscience, Institut Pasteur, Unité Neurobiologie Intégrative des Systèmes Cholinergiques, Paris Cedex 15, Paris, France
| | - Clotilde Lauro
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy
| | - Igor Branchi
- Section of Behavioural Neurosciences, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada.
| | - Laura Maggi
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| |
Collapse
|