1
|
Hsu CH, Hsu YY, Chang BM, Raffensperger K, Kadden M, Ton HT, Ette EA, Lin S, Brooks J, Burke MW, Lee YJ, Wang PC, Shoykhet M, Tu TW. StainAI: quantitative mapping of stained microglia and insights into brain-wide neuroinflammation and therapeutic effects in cardiac arrest. Commun Biol 2025; 8:462. [PMID: 40114030 PMCID: PMC11926354 DOI: 10.1038/s42003-025-07926-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Microglia, the brain's resident macrophages, participate in development and influence neuroinflammation, which is characteristic of multiple brain pathologies. Diverse insults cause microglia to alter their morphology from "resting" to "activated" shapes, which vary with stimulus type, brain location, and microenvironment. This morphologic diversity commonly restricts microglial analyses to specific regions and manual methods. We introduce StainAI, a deep learning tool that leverages 20x whole-slide immunohistochemistry images for rapid, high-throughput analysis of microglial morphology. StainAI maps microglia to a brain atlas, classifies their morphology, quantifies morphometric features, and computes an activation score for any region of interest. As a proof of principle, StainAI was applied to a rat model of pediatric asphyxial cardiac arrest, accurately classifying millions of microglia across multiple slices, surpassing current methods by orders of magnitude, and identifying both known and novel activation patterns. Extending its application to a non-human primate model of simian immunodeficiency virus infection further demonstrated its generalizability beyond rodent datasets, providing new insights into microglial responses across species. StainAI offers a scalable, high-throughput solution for microglial analysis from routine immunohistochemistry images, accelerating research in microglial biology and neuroinflammation.
Collapse
Affiliation(s)
- Chao-Hsiung Hsu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Yi-Yu Hsu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
- Miin Wu School of Computing, National Cheng Kung University, Tainan City, Taiwan
| | - Be-Ming Chang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Katherine Raffensperger
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, USA
| | - Micah Kadden
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, USA
- Pediatric Critical Care Medicine, Children's National Hospital, Washington, DC, USA
| | - Hoai T Ton
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, USA
| | - Essiet-Adidiong Ette
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Stephen Lin
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
| | - Janiya Brooks
- Department of Physiology and Biophysics, Howard University, Washington, DC, USA
| | - Mark W Burke
- Department of Physiology and Biophysics, Howard University, Washington, DC, USA
| | - Yih-Jing Lee
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Paul C Wang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA
- Department of Physics, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Michael Shoykhet
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC, USA
- Pediatric Critical Care Medicine, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Tsang-Wei Tu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA.
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
2
|
Nassrallah WB, Li HR, Irani L, Wijesinghe P, Hogg PW, Hui L, Oh J, Mackenzie IR, Hirsch-Reinshagen V, Hsiung GYR, Pham W, Lee S, Matsubara JA. 3-Dimensional morphological characterization of neuroretinal microglia in Alzheimer's disease via machine learning. Acta Neuropathol Commun 2024; 12:202. [PMID: 39719599 DOI: 10.1186/s40478-024-01898-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Alzheimer's Disease (AD) is a debilitating neurodegenerative disease that affects 47.5 million people worldwide. AD is characterised by the formation of plaques containing extracellular amyloid-β (Aβ) and neurofibrillary tangles composed of hyper-phosphorylated tau proteins (pTau). Aβ gradually accumulates in the brain up to 20 years before the clinical onset of dementia, making it a compelling candidate for early detection of AD. It has been shown that there is increased deposition of Aβs in AD patients' retinas. However, little is known about microglia's ability to function and clear Aβ within the retina of AD and control eyes. We labelled microglia with ionised calcium-binding adaptor molecule 1 (IBA-1) in AD and age-matched control donor retinas. We then used interactive machine learning to segment individual microglia in 3D. In the temporal mid-peripheral region, we found that the number of microglia was significantly lower in AD retinas compared to controls. Unexpectedly, the size of the microglia was significantly larger in the AD retinas compared to controls. We also labelled retinal microglia for Cluster of Differentiation 68 (CD68), a transmembrane glycoprotein expressed by cells in the monocyte lineage and a marker of phagocytic activity and activated microglia. The size of CD68 + cells was statistically different between AD and control microglial, with CD68 + cells being larger in AD. In contrast, there was no difference in either size or shape for CD68- microglia between the two groups, suggesting an important difference in the active states of CD68 + microglia in AD retina. There was also significantly increased CD68 immunoreactivity in individual microglia within the AD group. Overall, this study reveals unique differences in the size and activity of the retinal microglia, which may relate to their potential chronic activation due to increased levels of Aβs in the AD retina.
Collapse
Affiliation(s)
- Wissam B Nassrallah
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Hao Ran Li
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Lyden Irani
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Printha Wijesinghe
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Peter William Hogg
- Department of Cellular and Physiological Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Lucy Hui
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Jean Oh
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Ian R Mackenzie
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Veronica Hirsch-Reinshagen
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Ging-Yuek Robin Hsiung
- Division of Neurology, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Wellington Pham
- Vanderbilt University School of Medicine, Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Sieun Lee
- Simon Fraser University School of Engineering Science, Burnaby, BC, Canada
- Mental Health and Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada.
| |
Collapse
|
3
|
Lopez-Ortiz AO, Eyo UB. Astrocytes and microglia in the coordination of CNS development and homeostasis. J Neurochem 2024; 168:3599-3614. [PMID: 37985374 PMCID: PMC11102936 DOI: 10.1111/jnc.16006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/20/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023]
Abstract
Glia have emerged as important architects of central nervous system (CNS) development and maintenance. While traditionally glial contributions to CNS development and maintenance have been studied independently, there is growing evidence that either suggests or documents that glia may act in coordinated manners to effect developmental patterning and homeostatic functions in the CNS. In this review, we focus on astrocytes, the most abundant glia in the CNS, and microglia, the earliest glia to colonize the CNS highlighting research that documents either suggestive or established coordinated actions by these glial cells in various CNS processes including cell and/or debris clearance, neuronal survival and morphogenesis, synaptic maturation, and circuit function, angio-/vasculogenesis, myelination, and neurotransmission. Some molecular mechanisms underlying these processes that have been identified are also described. Throughout, we categorize the available evidence as either suggestive or established interactions between microglia and astrocytes in the regulation of the respective process and raise possible avenues for further research. We conclude indicating that a better understanding of coordinated astrocyte-microglial interactions in the developing and mature brain holds promise for developing effective therapies for brain pathologies where these processes are perturbed.
Collapse
Affiliation(s)
- Aída Oryza Lopez-Ortiz
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Ukpong B Eyo
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
4
|
Naughton SX, Yang EJ, Iqbal U, Trageser K, Charytonowicz D, Masieri S, Estill M, Wu H, Raval U, Lyu W, Wu QL, Shen L, Simon J, Sebra R, Pasinetti GM. Permethrin exposure primes neuroinflammatory stress response to drive depression-like behavior through microglial activation in a mouse model of Gulf War Illness. J Neuroinflammation 2024; 21:222. [PMID: 39272155 PMCID: PMC11396632 DOI: 10.1186/s12974-024-03215-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Gulf War Illness (GWI) is a chronic multisymptom disorder that affects approximately 25-32% of Gulf War veterans and is characterized by a number of symptoms such as cognitive impairment, psychiatric disturbances, chronic fatigue and gastrointestinal distress, among others. While the exact etiology of GWI is unknown, it is believed to have been caused by toxic exposures encountered during deployment in combination with other factors such as stress. In the present study we sought to evaluate the hypothesis that exposure to the toxin permethrin could prime neuroinflammatory stress response and elicit psychiatric symptoms associated with GWI. Specifically, we developed a mouse model of GWI, to evaluate the effects of chronic permethrin exposure followed by unpredictable stress. We found that subjecting mice to 14 days of chronic permethrin exposure followed by 7 days of unpredictable stress resulted in the development of depression-like behavior. This behavioral change coincided with distinct alterations in the microglia phenotype, indicating microglial activation in the hippocampus. We revealed that blocking microglial activation through Gi inhibitory DREADD receptors in microglia effectively prevented the behavioral change associated with permethrin and stress exposure. To elucidate the transcriptional networks impacted within distinct microglia populations linked to depression-like behavior in mice exposed to both permethrin and stress, we conducted a single-cell RNA sequencing analysis using 21,566 single nuclei collected from the hippocampus of mice. For bioinformatics, UniCell Deconvolve was a pre-trained, interpretable, deep learning model used to deconvolve cell type fractions and predict cell identity across spatial datasets. Our bioinformatics analysis identified significant alterations in permethrin exposure followed by stress-associated microglia population, notably pathways related to neuronal development, neuronal communication, and neuronal morphogenesis, all of which are associated with neural synaptic plasticity. Additionally, we observed permethrin exposure followed by stress-mediated changes in signal transduction, including modulation of chemical synaptic transmission, regulation of neurotransmitter receptors, and regulation of postsynaptic neurotransmitter receptor activity, a known contributor to the pathophysiology of depression in a subset of the hippocampal pyramidal neurons in CA3 subregions. Our findings tentatively suggest that permethrin may prime microglia towards a state of inflammatory activation that can be triggered by psychological stressors, resulting in depression-like behavior and alterations of neural plasticity. These findings underscore the significance of synergistic interactions between multi-causal factors associated with GWI.
Collapse
Affiliation(s)
- Sean X Naughton
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eun-Jeong Yang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Umar Iqbal
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kyle Trageser
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Charytonowicz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sibilla Masieri
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Molly Estill
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Henry Wu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Urdhva Raval
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Weiting Lyu
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, USA
| | - Qing-Li Wu
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, USA
| | - Li Shen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James Simon
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA.
| |
Collapse
|
5
|
Al-Musawi I, Dennis BH, Clowry GJ, LeBeau FEN. Evidence for prodromal changes in neuronal excitability and neuroinflammation in the hippocampus in young alpha-synuclein (A30P) transgenic mice. FRONTIERS IN DEMENTIA 2024; 3:1404841. [PMID: 39081599 PMCID: PMC11285622 DOI: 10.3389/frdem.2024.1404841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/20/2024] [Indexed: 08/02/2024]
Abstract
Introduction Neuronal hyperexcitability and neuroinflammation are thought to occur at early stages in a range of neurodegenerative diseases. Neuroinflammation, notably activation of microglia, has been identified as a potential prodromal marker of dementia with Lewy bodies (DLB). Using a transgenic mouse model of DLB that over-expresses human mutant (A30P) alpha-synuclein (hα-syn) we have investigated whether early neuroinflammation is evident in the hippocampus in young pre-symptomatic animals. Methods Previous studies have shown early hyperexcitability in the hippocampal CA3 region in male A30P mice at 2-4 months of age, therefore, in the current study we have immunostained this region for markers of neuronal activity (c-Fos), reactive astrocytes (glial fibrillary acidic protein, GFAP), microglia (ionizing calcium binding adapter protein 1, Iba-1) and reactive microglia (inducible nitric oxide synthase, iNOS). Results We found an interesting biphasic change in the expression of c-Fos in A30P mice with high expression at 1 month, consistent with early onset of hyperexcitability, but lower expression from 2-4 months in male A30P mice compared to wild-type (WT) controls, possibly indicating chronic hyperexcitability. Neuroinflammation was indicated by significant increases in the % area of GFAP and the number of Iba-1+ cells that expressed iNOS immunoreactivity in the CA3 region in 2-4 months A30P male mice compared to WT controls. A similar increase in % area of GFAP was observed in female A30P mice, however, the Iba-1 count was not different between female WT and A30P mice. In WT mice aged 2-4 months only 4.6% of Iba-1+ cells co-expressed iNOS. In contrast, in age matched A30P mice 87% of cells co-expressed Iba-1 and iNOS. Although there was no difference in GFAP immunoreactivity at 1 month, Iba-1/iNOS co-expression was also increased in a cohort of 1 month old A30P mice. Discussion Abnormal hα-syn expression in A30P mice caused early changes in network excitability, as indicated by c-Fos expression, and neuroinflammation which might contribute to disease progression.
Collapse
Affiliation(s)
| | | | | | - Fiona E. N. LeBeau
- Biosciences Institute and Centre for Transformative Neuroscience, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
6
|
Krejcová LV, Bento-Torres J, Diniz DG, Pereira A, Batista-de-Oliveira M, de Morais AACL, Mendes-da-Silva RF, Abadie-Guedes R, dos Santos ÂA, Lima DS, Guedes RCA, Picanço-Diniz CW. Unraveling the Influence of Litter Size, Maternal Care, Exercise, and Aging on Neurobehavioral Plasticity and Dentate Gyrus Microglia Dynamics in Male Rats. Brain Sci 2024; 14:497. [PMID: 38790475 PMCID: PMC11119659 DOI: 10.3390/brainsci14050497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
This study explores the multifaceted influence of litter size, maternal care, exercise, and aging on rats' neurobehavioral plasticity and dentate gyrus microglia dynamics. Body weight evolution revealed a progressive increase until maturity, followed by a decline during aging, with larger litters exhibiting lower weights initially. Notably, exercised rats from smaller litters displayed higher body weights during the mature and aged stages. The dentate gyrus volumes showed no significant differences among groups, except for aged sedentary rats from smaller litters, which exhibited a reduction. Maternal care varied significantly based on litter size, with large litter dams showing lower frequencies of caregiving behaviors. Behavioral assays highlighted the detrimental impact of a sedentary lifestyle and reduced maternal care/large litters on spatial memory, mitigated by exercise in aged rats from smaller litters. The microglial dynamics in the layers of dentate gyrus revealed age-related changes modulated by litter size and exercise. Exercise interventions mitigated microgliosis associated with aging, particularly in aged rats. These findings underscore the complex interplay between early-life experiences, exercise, microglial dynamics, and neurobehavioral outcomes during aging.
Collapse
Affiliation(s)
- Lane Viana Krejcová
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| | - João Bento-Torres
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| | - Daniel Guerreiro Diniz
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
- Postgraduate Program in Oncology and Medical Sciences, João de Barros Barreto Universitary Hospital, Federal University of Pará, Belém 66075-110, Pará, Brazil
- Electron Microscopy Laboratory, Evandro Chagas Institute, Belém 66093-020, Pará, Brazil
| | - Antonio Pereira
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| | - Manuella Batista-de-Oliveira
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | | | | | - Ricardo Abadie-Guedes
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Ângela Amâncio dos Santos
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Denise Sandrelly Lima
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Rubem Carlos Araujo Guedes
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Cristovam Wanderley Picanço-Diniz
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| |
Collapse
|
7
|
Rowson S, Bekhbat M, Kelly S, Hyer MM, Dyer S, Weinshenker D, Neigh G. Chronic adolescent stress alters GR-FKBP5 interactions in the hippocampus of adult female rats. Stress 2024; 27:2312467. [PMID: 38557197 PMCID: PMC11067065 DOI: 10.1080/10253890.2024.2312467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/25/2024] [Indexed: 04/04/2024] Open
Abstract
Chronic stress exposure during development can have lasting behavioral consequences that differ in males and females. More specifically, increased depressive behaviors in females, but not males, are observed in both humans and rodent models of chronic stress. Despite these known stress-induced outcomes, the molecular consequences of chronic adolescent stress in the adult brain are less clear. The stress hormone corticosterone activates the glucocorticoid receptor, and activity of the receptor is regulated through interactions with co-chaperones-such as the immunophilin FK506 binding proteins 5 (FKBP5). Previously, it has been reported that the adult stress response is modified by a history of chronic stress; therefore, the current study assessed the impact of chronic adolescent stress on the interactions of the glucocorticoid receptor (GR) with its regulatory co-chaperone FKBP5 in response to acute stress in adulthood. Although protein presence for FKBP5 did not differ by group, assessment of GR-FKBP5 interactions demonstrated that adult females with a history of chronic adolescent stress had elevated GR-FKBP5 interactions in the hippocampus following an acute stress challenge which could potentially contribute to a reduced translocation pattern given previous literature describing the impact of FKBP5 on GR activity. Interestingly, the altered co-chaperone interactions of the GR in the stressed female hippocampus were not coupled to an observable difference in transcription of GR-regulated genes. Together, these studies show that chronic adolescent stress causes lasting changes to co-chaperone interactions with the glucocorticoid receptor following stress exposure in adulthood and highlight the potential role that FKBP5 plays in these modifications. Understanding the long-term implications of adolescent stress exposure will provide a mechanistic framework to guide the development of interventions for adult disorders related to early life stress exposures.
Collapse
Affiliation(s)
- Sydney Rowson
- Molecular and Systems Pharmacology Graduate Program, Emory University, Atlanta, GA, USA
| | - Mandakh Bekhbat
- Neuroscience Graduate Program, Emory University, Atlanta, GA, USA
| | - Sean Kelly
- Department of Physiology, Emory University, Atlanta, GA, USA
| | - Molly M. Hyer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Samya Dyer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Gretchen Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
8
|
Jahanbakhshi H, Moghaddam MH, Sani M, Parvardeh S, Boroujeni ME, Vakili K, Fathi M, Azimi H, Mehranpour M, Abdollahifar MA, Ghafghazi S, Sadidi M, Aliaghaei A, Bayat AH, Peyvandi AA. The elderberry diet protection against intrahippocampal Aβ-induced memory dysfunction; the abrogated apoptosis and neuroinflammation. Toxicol Res (Camb) 2023; 12:1063-1076. [PMID: 38145093 PMCID: PMC10734613 DOI: 10.1093/toxres/tfad097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 12/26/2023] Open
Abstract
This study evaluates whether elderberry (EB) effectively decreases the inflammation and oxidative stress in the brain cells to reduce Aβ toxicity. In the Aβ + EB group, EB powder was added to rats' routine diet for eight consecutive weeks. Then, spatial memory, working memory, and long-term memory, were measured using the Morris water maze, T-maze, and passive avoidance test. Also, in this investigation immunohistopathology, distribution of hippocampal cells, and gene expression was carried out. Voronoi tessellation method was used to estimate the spatial distribution of the cells in the hippocampus. In addition to improving the memory functions of rats with Aβ toxicity, a reduction in astrogliosis and astrocytes process length and the number of branches and intersections distal to the soma was observed in their hippocampus compared to the control group. Further analysis indicated that the EB diet decreased the caspase-3 expression in the hippocampus of rats with Aβ toxicity. Also, EB protected hippocampal pyramidal neurons against Aβ toxicity and improved the spatial distribution of the hippocampal neurons. Moreover, EB decreased the expression of inflammatory and apoptotic genes. Overall, our study suggest that EB can be considered a potent modifier of astrocytes' reactivation and inflammatory responses.
Collapse
Affiliation(s)
- Hadiseh Jahanbakhshi
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mojtaba Sani
- Department of Educational Neuroscience, Aras International Campus, University of Tabriz, Tabriz, Iran
| | - Siavash Parvardeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Eskandarian Boroujeni
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Helia Azimi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Mehranpour
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shiva Ghafghazi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Sadidi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Hossein Bayat
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Asghar Peyvandi
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Lana D, Magni G, Landucci E, Wenk GL, Pellegrini-Giampietro DE, Giovannini MG. Phenomic Microglia Diversity as a Druggable Target in the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13668. [PMID: 37761971 PMCID: PMC10531074 DOI: 10.3390/ijms241813668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Phenomics, the complexity of microglia phenotypes and their related functions compels the continuous study of microglia in disease animal models to find druggable targets for neurodegenerative disorders. Activation of microglia was long considered detrimental for neuron survival, but more recently it has become apparent that the real scenario of microglia morphofunctional diversity is far more complex. In this review, we discuss the recent literature on the alterations in microglia phenomics in the hippocampus of animal models of normal brain aging, acute neuroinflammation, ischemia, and neurodegenerative disorders, such as AD. Microglia undergo phenomic changes consisting of transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. The classical subdivision of microglia into M1 and M2, two different, all-or-nothing states is too simplistic, and does not correspond to the variety of phenotypes recently discovered in the brain. We will discuss the phenomic modifications of microglia focusing not only on the differences in microglia reactivity in the diverse models of neurodegenerative disorders, but also among different areas of the brain. For instance, in contiguous and highly interconnected regions of the rat hippocampus, microglia show a differential, finely regulated, and region-specific reactivity, demonstrating that microglia responses are not uniform, but vary significantly from area to area in response to insults. It is of great interest to verify whether the differences in microglia reactivity may explain the differential susceptibility of different brain areas to insults, and particularly the higher sensitivity of CA1 pyramidal neurons to inflammatory stimuli. Understanding the spatiotemporal heterogeneity of microglia phenomics in health and disease is of paramount importance to find new druggable targets for the development of novel microglia-targeted therapies in different CNS disorders. This will allow interventions in three different ways: (i) by suppressing the pro-inflammatory properties of microglia to limit the deleterious effect of their activation; (ii) by modulating microglia phenotypic change to favor anti-inflammatory properties; (iii) by influencing microglia priming early in the disease process.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Florence, Italy;
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Gary L. Wenk
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA;
| | - Domenico Edoardo Pellegrini-Giampietro
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| |
Collapse
|
10
|
Stephen TL, Korobkova L, Breningstall B, Nguyen K, Mehta S, Pachicano M, Jones KT, Hawes D, Cabeen RP, Bienkowski MS. Machine Learning Classification of Alzheimer's Disease Pathology Reveals Diffuse Amyloid as a Major Predictor of Cognitive Impairment in Human Hippocampal Subregions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.31.543117. [PMID: 37333119 PMCID: PMC10274752 DOI: 10.1101/2023.05.31.543117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Analyzing Alzheimer's disease (AD) pathology within anatomical subregions is a significant challenge, often carried out by pathologists using a standardized, semi-quantitative approach. To augment traditional methods, a high-throughput, high-resolution pipeline was created to classify the distribution of AD pathology within hippocampal subregions. USC ADRC post-mortem tissue sections from 51 patients were stained with 4G8 for amyloid, Gallyas for neurofibrillary tangles (NFTs) and Iba1 for microglia. Machine learning (ML) techniques were utilized to identify and classify amyloid pathology (dense, diffuse and APP (amyloid precursor protein)), NFTs, neuritic plaques and microglia. These classifications were overlaid within manually segmented regions (aligned with the Allen Human Brain Atlas) to create detailed pathology maps. Cases were separated into low, intermediate, or high AD stages. Further data extraction enabled quantification of plaque size and pathology density alongside ApoE genotype, sex, and cognitive status. Our findings revealed that the increase in pathology burden across AD stages was driven mainly by diffuse amyloid. The pre and para-subiculum had the highest levels of diffuse amyloid while NFTs were highest in the A36 region in high AD cases. Moreover, different pathology types had distinct trajectories across disease stages. In a subset of AD cases, microglia were elevated in intermediate and high compared to low AD. Microglia also correlated with amyloid pathology in the Dentate Gyrus. The size of dense plaques, which may represent microglial function, was lower in ApoE4 carriers. In addition, individuals with memory impairment had higher levels of both dense and diffuse amyloid. Taken together, our findings integrating ML classification approaches with anatomical segmentation maps provide new insights on the complexity of disease pathology in AD progression. Specifically, we identified diffuse amyloid pathology as being a major driver of AD in our cohort, regions of interest and microglial responses that might advance AD diagnosis and treatment.
Collapse
|
11
|
Costello A, Linning-Duffy K, Vandenbrook C, Lonstein JS, Yan L. Daytime Light Deficiency Leads to Sex- and Brain Region-Specific Neuroinflammatory Responses in a Diurnal Rodent. Cell Mol Neurobiol 2023; 43:1369-1384. [PMID: 35864429 PMCID: PMC10635710 DOI: 10.1007/s10571-022-01256-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/07/2022] [Indexed: 11/30/2022]
Abstract
Seasonal changes in peripheral inflammation are well documented in both humans and animal models, but seasonal changes in neuroinflammation, especially the impact of seasonal lighting environment on neuroinflammation remain unclear. To address this question, the present study examined the effects of environmental lighting conditions on neuroinflammation in a diurnal rodent model, Nile grass rats (Arvicanthis niloticus). Male and female grass rats were housed in either bright (brLD) or dim (dimLD) light during the day to simulate a summer or winter light condition, respectively. After 4 weeks, microglia markers Iba-1 and CD11b, as well as pro-inflammatory cytokines TNF-α and IL-6, were examined in the anterior cingulate cortex (ACC), basolateral amygdala (BLA), and dorsal hippocampus (dHipp). The results revealed that winter-like dim light during the day leads to indicators of increased neuroinflammation in a brain site- and sex-specific manner. Specifically, relatively few changes in the neuroinflammatory markers were observed in the ACC, while numerous changes were found in the BLA and dHipp. In the BLA, winter-like dimLD resulted in hyper-ramified microglia morphology and increased expression of the pro-inflammatory cytokine IL-6, but only in males. In the dHipp, dimLD led to a higher number and hyper-ramified morphology of microglia as well as increased expression of CD11b and TNF-α, but only in females. Neuroinflammatory state is thus influenced by environmental light, differently in males and females, and could play a role in sex differences in the prevalence and symptoms of psychiatric or neurological disorders that are influenced by season or other environmental light conditions. Diurnal Nile grass rats were housed under bright or dim light during the day for 4 weeks, simulating seasonal fluctuations in daytime lighting environment. Dim light housing resulted in hyper-ramified morphology of microglia (scale bar, 15 μm) and altered expression of pro-inflammatory cytokines (TNF-α) in a sex- and brain region-specific manner.
Collapse
Affiliation(s)
- Allison Costello
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA
| | - Katrina Linning-Duffy
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA
| | - Carleigh Vandenbrook
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA
| | - Joseph S Lonstein
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Lily Yan
- Behavioral Neuroscience Program, Department of Psychology, Michigan State University, 766, Service Road, East Lansing, MI, 48824, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
12
|
De Felice E, Gonçalves de Andrade E, Golia MT, González Ibáñez F, Khakpour M, Di Castro MA, Garofalo S, Di Pietro E, Benatti C, Brunello N, Tascedda F, Kaminska B, Limatola C, Ragozzino D, Tremblay ME, Alboni S, Maggi L. Microglial diversity along the hippocampal longitudinal axis impacts synaptic plasticity in adult male mice under homeostatic conditions. J Neuroinflammation 2022; 19:292. [PMID: 36482444 PMCID: PMC9730634 DOI: 10.1186/s12974-022-02655-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is a plastic brain area that shows functional segregation along its longitudinal axis, reflected by a higher level of long-term potentiation (LTP) in the CA1 region of the dorsal hippocampus (DH) compared to the ventral hippocampus (VH), but the mechanisms underlying this difference remain elusive. Numerous studies have highlighted the importance of microglia-neuronal communication in modulating synaptic transmission and hippocampal plasticity, although its role in physiological contexts is still largely unknown. We characterized in depth the features of microglia in the two hippocampal poles and investigated their contribution to CA1 plasticity under physiological conditions. We unveiled the influence of microglia in differentially modulating the amplitude of LTP in the DH and VH, showing that minocycline or PLX5622 treatment reduced LTP amplitude in the DH, while increasing it in the VH. This was recapitulated in Cx3cr1 knockout mice, indicating that microglia have a key role in setting the conditions for plasticity processes in a region-specific manner, and that the CX3CL1-CX3CR1 pathway is a key element in determining the basal level of CA1 LTP in the two regions. The observed LTP differences at the two poles were associated with transcriptional changes in the expression of genes encoding for Il-1, Tnf-α, Il-6, and Bdnf, essential players of neuronal plasticity. Furthermore, microglia in the CA1 SR region showed an increase in soma and a more extensive arborization, an increased prevalence of immature lysosomes accompanied by an elevation in mRNA expression of phagocytic markers Mertk and Cd68 and a surge in the expression of microglial outward K+ currents in the VH compared to DH, suggesting a distinct basal phenotypic state of microglia across the two hippocampal poles. Overall, we characterized the molecular, morphological, ultrastructural, and functional profile of microglia at the two poles, suggesting that modifications in hippocampal subregions related to different microglial statuses can contribute to dissect the phenotypical aspects of many diseases in which microglia are known to be involved.
Collapse
Affiliation(s)
- E. De Felice
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Gonçalves de Andrade
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. T. Golia
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - F. González Ibáñez
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - M. Khakpour
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada
| | - M. A. Di Castro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - S. Garofalo
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - E. Di Pietro
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - C. Benatti
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - N. Brunello
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - F. Tascedda
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - B. Kaminska
- grid.419305.a0000 0001 1943 2944Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - C. Limatola
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur, Sapienza University, Rome, Italy
| | - D. Ragozzino
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy ,grid.417778.a0000 0001 0692 3437Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - M. E. Tremblay
- grid.143640.40000 0004 1936 9465Division of Medical Sciences, University of Victoria, Victoria, Canada ,grid.411081.d0000 0000 9471 1794Faculté de Médecine and Centre de Recherche, CHU de Québec-Université Laval, Quebec, Canada
| | - S. Alboni
- grid.7548.e0000000121697570Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy ,grid.7548.e0000000121697570Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - L. Maggi
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| |
Collapse
|
13
|
Proteomic Signature and mRNA Expression in Hippocampus of SAMP8 and SAMR1 Mice during Aging. Int J Mol Sci 2022; 23:ijms232315097. [PMID: 36499421 PMCID: PMC9740614 DOI: 10.3390/ijms232315097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Aging is a complex process often accompanied by cognitive decline that represents a risk factor for many neurodegenerative disorders including Alzheimer's and Parkinson's disease. The molecular mechanisms involved in age-related cognitive decline are not yet fully understood, although increased neuroinflammation is considered to play a significant role. In this study, we characterized a proteomic view of the hippocampus of the senescence-accelerated mouse prone-8 (SAMP8), a model of enhanced senescence, in comparison with the senescence-accelerated-resistant mouse (SAMR1), a model of normal aging. We additionally investigated inflammatory cytokines and cholinergic components gene expression during aging in the mouse brain tissues. Proteomic data defined the expression of key proteins involved in metabolic and cellular processes in neuronal and glial cells of the hippocampus. Gene Ontology revealed that most of the differentially expressed proteins are involved in the cytoskeleton and cell motility regulation. Molecular analysis results showed that both inflammatory cytokines and cholinergic components are differentially expressed during aging, with a downward trend of cholinergic receptors and esterase enzymes expression, in contrast to an upward trend of inflammatory cytokines in the hippocampus of SAMP8. Together, our results support the important role of the cholinergic and cytokine systems in the aging of the murine brain.
Collapse
|
14
|
McCamy KM, Rees KA, Winzer-Serhan UH. Peripheral immune challenges elicit differential up-regulation of hippocampal cytokine and chemokine mRNA expression in a mouse model of the 15q13.3 microdeletion syndrome. Cytokine 2022; 159:156005. [PMID: 36084604 DOI: 10.1016/j.cyto.2022.156005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/06/2022] [Accepted: 08/05/2022] [Indexed: 11/03/2022]
Abstract
The human heterozygous 15q13.3 microdeletion is associated with neuropathological disorders, most prominently with epilepsy and intellectual disability. The 1.5 Mb deletion encompasses six genes (FAN1 [MTMR15], MTMR10, TRPM1, KLF13, OTUD7A, and CHRNA7); all but one (TRPM1) are expressed in the brain. The 15q13.3 microdeletion causes highly variable neurological symptoms, and confounding factors may contribute to a more severe phenotype. CHRNA7 and KLF13 are involved in immune system regulation and altered immune responses may contribute to neurological deficits. We used the Df[h15q13]/+ transgenic mouse model with a heterozygous deletion of the orthologous region (Het) to test the hypothesis that the microdeletion increases innate immune responses compared to wild type (WT). Male and female mice were acutely challenged with the bacteriomimetic lipopolysaccharide (LPS, 0.1 mg/kg, i.p.) or the viral mimetic polyinosinic:polycytidylic acid (Poly(I:C), 5 mg/kg). Hippocampal mRNA expression of pro-inflammatory cytokines and chemokines were determined three hours after injection using quantitative PCR analysis. In controls, expression was not affected by sex or genotype. LPS and Poly(I:C) resulted in significantly increased hippocampal expression of cytokines, chemokines, and interferon-γ (IFNγ), with more robust increases for TNF-α, IL-6, IL-1β, CXCL1, and CCL2 by LPS, higher induction of IFNγ by Poly(I:C), and similar increases of CCL4 and CCL5 by both agents. Generally, Hets exhibited stronger responses than WT mice, and significant effects of genotype or genotype × treatment interactions were detected for CXCL1 and CCL5, and IL-6, IL-1β, and CCL4, respectively, after LPS. Sex differences were detected for some targets. LPS but not Poly(I:C), reduced overnight burrowing independent of sex or genotype, suggesting that LPS induced sickness behavior. Thus, mice carrying the microdeletion have an increased innate immune response following a LPS challenge, but further studies will have to determine the extent and mechanisms of altered immune activation and subsequent contributions to 15q13.3 microdeletion associated deficits.
Collapse
Affiliation(s)
- Kristin M McCamy
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States
| | - Katherine A Rees
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States
| | - Ursula H Winzer-Serhan
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States.
| |
Collapse
|
15
|
Zilli J, Schänzer A, Büttner K, Kressin M, Schmidt MJ. Quantitative and qualitative evaluation of the hippocampal cytoarchitecture in adult cats with regard to the pathological diagnosis of hippocampal sclerosis. PLoS One 2022; 17:e0268010. [PMID: 35560321 PMCID: PMC9106214 DOI: 10.1371/journal.pone.0268010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/20/2022] [Indexed: 11/18/2022] Open
Abstract
Cats are known to be affected by hippocampal sclerosis, potentially causing antiseizure drug(s) resistance. In order to lay the foundation for a standardized, systematic classification and diagnosis of this pathology in cats, this prospective study aimed at evaluating normal reference values of cellular densities and the cytoarchitecture of the feline hippocampus. Three transverse sections (head, body and tail) of each left hippocampus were obtained from 17 non-epileptic cats of different brachycephalic and mesocephalic breeds and age classes (range: 3–17 years). Histological (hematoxylin and eosin, Nissl) and immunohistochemical (NeuN, GFAP) staining was performed to investigate neuron and astroglial cell populations, as well as the layer thickness of the pyramidal cell layer and granule cell layer. Significant differences in neuronal density (in CA2-CA4 and the granule cell layer) and layer thickness (in CA1-CA3 and the granule cell layer) were evidenced throughout the longitudinal hippocampal axis (p<0.05); on the other hand, the astrocyte density did not differ. Moreover, reference ranges were defined for these parameters in the pyramidal cell layer and in the granule cell layer. The findings did not differ according to breed or age. In veterinary medicine these parameters have not been evaluated in cats so far. As surgical treatment may become a therapeutic option for cats with temporal lobe epilepsy, estimating normal values of the hippocampal cytoarchitecture will help in the standardized histopathological examination of resected hippocampal specimens to reach a diagnosis of hippocampal sclerosis.
Collapse
Affiliation(s)
- Jessica Zilli
- Department of Veterinary Clinical Sciences, Small Animal Clinic, Justus-Liebig-University, Giessen, Hessen, Germany
- * E-mail:
| | - Anne Schänzer
- Institute of Neuropathology, Justus-Liebig-University, Giessen, Hessen, Germany
| | - Kathrin Büttner
- Institute for Biomathematics, Justus-Liebig-University, Giessen, Hessen, Germany
| | - Monika Kressin
- Institute for Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University, Giessen, Hessen, Germany
| | - Martin J. Schmidt
- Department of Veterinary Clinical Sciences, Small Animal Clinic, Justus-Liebig-University, Giessen, Hessen, Germany
| |
Collapse
|
16
|
Tang JJ, Huang LF, Deng JL, Wang YM, Guo C, Peng XN, Liu Z, Gao JM. Cognitive enhancement and neuroprotective effects of OABL, a sesquiterpene lactone in 5xFAD Alzheimer's disease mice model. Redox Biol 2022; 50:102229. [PMID: 35026701 PMCID: PMC8760418 DOI: 10.1016/j.redox.2022.102229] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 12/30/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease in which oxidative stress and neuroinflammation were demonstrated to be associated with neuronal loss and cognitive deficits. However, there are still no specific treatments that can prevent the progression of AD. In this study, a screening of anti-inflammatory hits from 4207 natural compounds of two different molecular libraries indicated 1,6-O,O-diacetylbritannilactone (OABL), a 1,10-seco-eudesmane sesquiterpene lactone isolated from the herb Inula britannica L., exhibited strong anti-inflammatory activity in vitro as well as favorable BBB penetration property. OABL reduced LPS-induced neuroinflammation in BV-2 microglial cells as assessed by effects on the levels of inflammatory mediators including NO, PGE2, TNF-α, iNOS, and COX-2, as well as the translocation of NF-κB. Besides, OABL also exhibited pronounced neuroprotective effects against oxytosis and ferroptosis in the rat pheochromocytoma PC12 cell line. For in vivo research, OABL (20 mg/kg B.W., i.p.) for 21 d attenuated the impairments in cognitive function observed in 6-month-old 5xFAD mice, as assessed with the Morris water maze test. OABL restored neuronal damage and postsynaptic density protein 95 (PSD95) expression in the hippocampus. OABL also significantly reduced the accumulation of amyloid plaques, the Aβ expression, the phosphorylation of Tau protein, and the expression of BACE1 in AD mice brain. In addition, OABL attenuated the overactivation of microglia and astrocytes by suppressing the expressions of inflammatory cytokines, and increased glutathione (GSH) and reduced malondialdehyde (MDA) and super oxide dismutase (SOD) levels in the 5xFAD mice brain. In conclusion, these results highlight the beneficial effects of the natural product OABL as a novel treatment with potential application for drug discovery in AD due to its pharmacological profile.
Collapse
Affiliation(s)
- Jiang-Jiang Tang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shaanxi, PR China.
| | - Lan-Fang Huang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Jia-Le Deng
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Yi-Meng Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Cong Guo
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Xiao-Na Peng
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, Shaanxi, PR China.
| | - Jin-Ming Gao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, Shaanxi, PR China.
| |
Collapse
|
17
|
Ohgomori T, Jinno S. Potential Involvement of Keratan Sulfate in the Heterogeneity of Microglia. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2038.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Tomohiro Ohgomori
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University
| |
Collapse
|
18
|
Ohgomori T, Jinno S. Potential Involvement of Keratan Sulfate in the Heterogeneity of Microglia. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2038.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Tomohiro Ohgomori
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University
| |
Collapse
|
19
|
Tyrtyshnaia A, Konovalova S, Bondar A, Ermolenko E, Sultanov R, Manzhulo I. Anti-Inflammatory Activity of N-Docosahexaenoylethanolamine and N-Eicosapentaenoylethanolamine in a Mouse Model of Lipopolysaccharide-Induced Neuroinflammation. Int J Mol Sci 2021; 22:ijms221910728. [PMID: 34639071 PMCID: PMC8509568 DOI: 10.3390/ijms221910728] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/16/2022] Open
Abstract
The search for methods of cognitive impairment treatment and prevention in neurological and neurodegenerative diseases is an urgent task of modern neurobiology. It is now known that various diseases, accompanied by dementia, exhibit a pronounced neuroinflammation. Considering the significant docosahexaenoic and eicosapentaenoic polyunsaturated fatty acids' therapeutic potential, we decided to investigate and compare anti-inflammatory activity of their N-acylethanolamine derivatives. As a result, we found that both N-docosahexaenoylethanolamine (synaptamide) and N-eicosapentaenoylethanolamine (EPEA) prevents an LPS-mediated increase in the proinflammatory cytokines TNF-α and IL-6 production in the SIM-A9 microglia culture. In an in vivo experiment, synaptamide reversed an increase in LPS-mediated hippocampal TNF-α and IL-1β, but EPEA did not. However, both compounds contributed to the microglia polarization towards the M2-phenotype. Synaptamide, rather than EPEA, inhibited the Iba-1-positive microglia staining area increase. However, both synaptamide and EPEA prevented the LPS-mediated astrogliosis. A study of BDNF immunoreactivity showed that synaptamide, but not EPEA, reversed an LPS-mediated decrease in BDNF production. Despite the more pronounced anti-inflammatory activity of synaptamide, both compounds were effective in maintaining a normal level of hippocampal long-term potentiation in neuroinflammation. The results indicate a high therapeutic potential for both compounds. However, some tests have shown higher activity of synaptamide compared to EPEA.
Collapse
|
20
|
Maternal separation leads to regional hippocampal microglial activation and alters the behavior in the adolescence in a sex-specific manner. Brain Behav Immun Health 2021; 9:100142. [PMID: 34589889 PMCID: PMC8474514 DOI: 10.1016/j.bbih.2020.100142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/04/2020] [Accepted: 09/13/2020] [Indexed: 12/31/2022] Open
Abstract
Early life adversities during childhood (such as maltreatment, abuse, neglect, or parental deprivation) may increase the vulnerability to cognitive disturbances and emotional disorders in both, adolescence and adulthood. Maternal separation (MS) is a widely used model to study stress-related changes in brain and behavior in rodents. In this study, we investigated the effect of MS (postnatal day 2–14, 3 h/day) in both, female and male adolescent mice. Specifically, we evaluated (i) the spatial working memory, anxiety and depressive-like behavior, (ii) the hippocampal synaptic gene expression, and (iii) the hippocampal neuroinflammatory response. Our results show that MS significantly increased depressive-like behavior in adolescent female mice and altered the spatial memory in adolescent male mice. In addition, MS led to decreased expression of genes related to synaptic function (5ht6r, Synaptophysin, and Cox-2) and induced an exacerbated microglial activation in dentate gyrus (DG), CA1, and CA3. However, while the levels of hippocampal inflammatory cytokines were not modified by MS, they did follow a sex-specific expression in adolescent mice. Taken together, our results suggest that MS induces long-term changes in hippocampal microglia and synaptic gene expression, alters the spatial memory, and induces depressive-like behavior in the adolescent mice, in a sex-specific manner. In wildtype adolescent mice (6 weeks of age):Maternal separation alters spatial working memory in males and induces depressive-like behavior in females. Maternal separation changes hippocampal synaptic gene expression. Maternal separation activates microglia in dentate gyrus, CA1, and CA3 but does not affect hippocampal cytokine levels. However, males present higher levels of cytokines compared to females.
Collapse
|
21
|
Manabe T, Rácz I, Schwartz S, Oberle L, Santarelli F, Emmrich JV, Neher JJ, Heneka MT. Systemic inflammation induced the delayed reduction of excitatory synapses in the CA3 during ageing. J Neurochem 2021; 159:525-542. [PMID: 34379806 DOI: 10.1111/jnc.15491] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/27/2021] [Accepted: 08/09/2021] [Indexed: 11/29/2022]
Abstract
Sepsis-associated encephalopathy (SAE) represents diverse cerebral dysfunctions in response to pathogen-induced systemic inflammation. Peripheral exposure to lipopolysaccharide (LPS), a component of the gram-negative bacterial cell wall, has been extensively used to model systemic inflammation. Our previous studies suggested that LPS led to hippocampal neuron death and synaptic destruction in vivo. However, the underlying roles of activated microglia in these neuronal changes remained unclear. Here, LPS from two different bacterial strains (Salmonella enterica or E. coli) were compared and injected in 14- to 16-month-old mice and evaluated for neuroinflammation and neuronal integrity in the hippocampus at 7 or 63 days post-injection (dpi). LPS injection resulted in persistent neuroinflammation lasting for seven days and a subsequent normalisation by 63 dpi. Of note, increases in proinflammatory cytokines, microglial morphology and microglial mean lysosome volume were more pronounced after E. coli LPS injection than Salmonella LPS at 7 dpi. While inhibitory synaptic puncta density remained normal, excitatory synaptic puncta were locally reduced in the CA3 region of the hippocampus at 63 dpi. Finally, we provide evidence that excitatory synapses coated with complement factor 3 (C3) decreased between 7 dpi and 63 dpi. Although we did not find an increase of synaptic pruning by microglia, it is plausible that microglia recognised and eliminated these C3-tagged synapses between the two time-points of investigation. Since a region-specific decline of CA3 synapses has previously been reported during normal ageing, we postulate that systemic inflammation may have accelerated or worsened the CA3 synaptic changes in the ageing brain.
Collapse
Affiliation(s)
- Tatsuya Manabe
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
| | - Ildikó Rácz
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
| | - Stephanie Schwartz
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
| | - Linda Oberle
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
| | | | - Julius V Emmrich
- Department of Neurology and Department of Experimental Neurology, Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, Berlin, 10117, Germany
| | - Jonas J Neher
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn Medical Center, Venusberg-Campus 1, 53127, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany.,Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, 01655, USA
| |
Collapse
|
22
|
Schilling S, Chausse B, Dikmen HO, Almouhanna F, Hollnagel JO, Lewen A, Kann O. TLR2- and TLR3-activated microglia induce different levels of neuronal network dysfunction in a context-dependent manner. Brain Behav Immun 2021; 96:80-91. [PMID: 34015428 DOI: 10.1016/j.bbi.2021.05.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/11/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023] Open
Abstract
Recognition of pathogen- or damage-associated molecular patterns (PAMPs, DAMPs) by innate Toll-like receptors (TLRs) is central to the activation of microglia (brain macrophages) in many CNS diseases. Notably, TLR-mediated microglial activation is complex and modulated by additional exogenous and endogenous immunological signals. The impact of different microglial reactive phenotypes on electrical activity and neurotransmission is widely unknown, however. We explored the effects of TLR ligands on microglia and neuronal network function in rat organotypic hippocampal slice cultures (in situ), i.e., postnatal cortical tissue lacking adaptive immunity. Single exposure of slice cultures to TLR2 or TLR3 ligands [PGN, poly(I:C)] for 2-3 days induced moderate microglial activation featuring IL-6 and TNF-α release and only mild alterations of fast neuronal gamma band oscillations (30-70 Hz) that are fundamental to higher cognitive functions, such as perception, memory and behavior. Paired exposure to TLR3/TLR2 or TLR3/TLR4 ligands (LPS) induced nitric oxide (NO) release, enhanced TNF-α release, and associated with advanced network dysfunction, including slowing to the beta frequency band (12-30 Hz) and neural bursts (hyperexcitability). Paired exposure to a TLR ligand and the leukocyte cytokine IFN-γ enhanced NO release and associated with severe network dysfunction, albeit sensitive parvalbumin- and somatostatin-positive inhibitory interneurons were preserved. Notably, the neuronal disturbance was prevented by either microglial depletion or pharmacological inhibition of oxidant-producing enzymes, inducible NO synthase (iNOS) and NADPH oxidase. In conclusion, TLR-activated microglia can induce different levels of neuronal network dysfunction, in which severe dysfunction is mainly caused by reactive oxygen and nitrogen species rather than proinflammatory cytokines. Our findings provide a mechanistic insight into microglial activation and functional neuronal network impairment, with relevance to neuroinflammation and neurodegeneration observed in, e.g., meningoencephalitis, multiple sclerosis and Alzheimer's disease.
Collapse
Affiliation(s)
- Simone Schilling
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Bruno Chausse
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Hasan Onur Dikmen
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Fadi Almouhanna
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Jan-Oliver Hollnagel
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Andrea Lewen
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, D-69120 Heidelberg, Germany.
| |
Collapse
|
23
|
Zetter MA, Hernández VS, Roque A, Hernández-Pérez OR, Gómora MJ, Ruiz-Velasco S, Eiden LE, Zhang L. Microglial synaptic pruning on axon initial segment spines of dentate granule cells: Sexually dimorphic effects of early-life stress and consequences for adult fear response. J Neuroendocrinol 2021; 33:e12969. [PMID: 33890333 DOI: 10.1111/jne.12969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/22/2022]
Abstract
Axon initial segments (AIS) of dentate granule cells in the hippocampus exhibit prominent spines (AISS) during early development that are associated with microglial contacts. In the present study, we investigated whether developmental changes in AISS could be modified by early-life stress (ELS), specifically neonatal maternal separation (MS), through stress hormones and microglial activation and examined the potential behavioural consequences. We examined AISS at postnatal day (PND)5, 15 and 50, using Golgi-Cox staining and anatomical analysis. Neurone-microglial interaction was assessed using antibodies against ankyrin-G, PSD-95 and Iba1, for AIS, AISS and microglia visualisation, respectively, in normally reared and neonatal maternally separated male and female rats. We observed a higher density of AISS in ELS rats at both PND15 and PND50 compared to controls. Effects were more pronounced in females than males. AIS-associated microglia in ELS rats showed a hyper-ramified morphology and less co-localisation with PSD-95 compared to controls at PND15. ELS-associated alteration in microglial morphology and synaptic pruning was mimicked by treatment of acute hippocampal slices of normally reared rats with vasopressin. ELS rats exhibited increased freezing behaviour during auditory fear memory testing, which was more pronounced in female subjects and corresponded with increased Fos expression in dorsal and ventral dentate granule cells. Thus, microglial synaptic pruning in dentate AIS of hippocampus is influenced by ELS, with demonstrable sex bias regarding its anatomical characteristics and subsequent fear-induced defensive behaviours.
Collapse
Affiliation(s)
- Mario A Zetter
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Vito S Hernández
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Angélica Roque
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Oscar R Hernández-Pérez
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - María J Gómora
- Department of Embryology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Silvia Ruiz-Velasco
- Department of Probability and Statistics, Applied Mathematics and Systems Research Institute, National Autonomous University of Mexico, Mexico City, Mexico
| | - Lee E Eiden
- Section on Molecular Neuroscience, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Limei Zhang
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
24
|
Ohgomori T, Iinuma K, Yamada J, Jinno S. A unique subtype of ramified microglia associated with synapses in the rat hippocampus. Eur J Neurosci 2021; 54:4740-4754. [PMID: 34110047 DOI: 10.1111/ejn.15330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/13/2022]
Abstract
To date, a number of studies have reported the heterogeneity of activated microglia. However, there is increasing evidence suggests that ramified, so-called resting, microglia may also be heterogeneous, and they may play diverse roles in normal brain homeostasis. Here, we found that both 5D4 keratan sulfate epitope-positive (5D4+ ) and 5D4-negative (5D4- ) microglia coexisted in the hippocampus of normal rats, while all microglia were negative for the 5D4 epitope in the hippocampus of normal mice. We thus aimed to determine the potential heterogeneity of microglia related to the 5D4 epitope in the normal rat hippocampus. The optical disector analysis showed that the densities of 5D4+ microglia were higher in the stratum oriens of the CA3 region than in other layers and regions. Although both 5D4+ and 5D4- microglia exhibited a ramified morphology, the three-dimensional reconstruction analysis showed that the node numbers, end numbers, and complexity of processes were higher in 5D4+ than in 5D4- microglia. The linear discriminant analysis showed that 5D4+ and 5D4- microglia can be classified into distinct morphometric subtypes. The ratios of contact between synaptic boutons and microglial processes were higher in 5D4+ than in 5D4- microglia. The gene expressions of pro-inflammatory cytokine interleukin-1β and purinergic receptor P2Y12 (P2Y12 R) were higher in 5D4+ than in 5D4- microglia. Together, these results indicate that at least two different subtypes of ramified microglia coexist in the normal rat hippocampus and also suggest that 5D4+ microglia may represent a unique subtype associated with synapses.
Collapse
Affiliation(s)
- Tomohiro Ohgomori
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, Kaizuka, Japan
| | - Kyoko Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
25
|
Li Y, Tang W, Guo M. The Cell as Matter: Connecting Molecular Biology to Cellular Functions. MATTER 2021; 4:1863-1891. [PMID: 35495565 PMCID: PMC9053450 DOI: 10.1016/j.matt.2021.03.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Viewing cell as matter to understand the intracellular biomolecular processes and multicellular tissue behavior represents an emerging research area at the interface of physics and biology. Cellular material displays various physical and mechanical properties, which can strongly affect both intracellular and multicellular biological events. This review provides a summary of how cells, as matter, connect molecular biology to cellular and multicellular scale functions. As an impact in molecular biology, we review recent progresses in utilizing cellular material properties to direct cell fate decisions in the communities of immune cells, neurons, stem cells, and cancer cells. Finally, we provide an outlook on how to integrate cellular material properties in developing biophysical methods for engineered living systems, regenerative medicine, and disease treatments.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wenhui Tang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
26
|
Lacto-N-fucopentaose-III (LNFPIII) ameliorates acute aberrations in hippocampal synaptic transmission in a Gulf War Illness animal model. Brain Res 2021; 1766:147513. [PMID: 33961896 DOI: 10.1016/j.brainres.2021.147513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 02/04/2023]
Abstract
Approximately one-third of Persian Gulf War veterans are afflicted by Gulf War Illness (GWI), a chronic multisymptom condition that fundamentally presents with cognitive deficits (i.e., learning and memory impairments) and neuroimmune dysfunction (i.e., inflammation). Factors associated with GWI include overexposures to neurotoxic pesticides and nerve agent prophylactics such as permethrin (PM) and pyridostigmine bromide (PB), respectively. GWI-related neurological impairments associated with PB-PM overexposures have been recapitulated in animal models; however, there is a paucity of studies assessing PB-PM-related aberrations in hippocampal synaptic plasticity and transmission that may underlie behavioral impairments. Importantly, FDA-approved neuroactive treatments are currently unavailable for GWI. In the present study, we assessed the efficacy of an immunomodulatory therapeutic, lacto-N-fucopentaose-III (LNFPIII), on ameliorating acute effects of in vivo PB-PM exposure on synaptic plasticity and transmission as well as trophic factor/cytokine expression along the hippocampal dorsoventral axis. PB-PM exposure resulted in hippocampal synaptic transmission deficits 48 h post-exposure, a response that was ameliorated by LNFPIII coadministration, particularly in the dorsal hippocampus (dH). LNFPIII coadministration also enhanced synaptic transmission in the dH and the ventral hippocampus (vH). Notably, LNFPIII coadministration elevated long-term potentiation in the dH. Further, PB-PM exposure and LNFPIII coadministration uniquely altered key inflammatory cytokine and trophic factor production in the dH and the vH. Collectively, these findings demonstrate that PB-PM exposure impaired hippocampal synaptic responses 48 h post-exposure, impairments that differentially manifested along the dorsoventral axis. Importantly, LNFPIII ameliorated GWI-related electrophysiological deficits, a beneficial effect indicating the potential efficacy of LNFPIII for treating GWI.
Collapse
|
27
|
Carrera J, Tomberlin J, Kurtz J, Karakaya E, Bostanciklioglu M, Albayram O. Endocannabinoid Signaling for GABAergic-Microglia (Mis)Communication in the Brain Aging. Front Neurosci 2021; 14:606808. [PMID: 33613174 PMCID: PMC7887316 DOI: 10.3389/fnins.2020.606808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/28/2020] [Indexed: 12/11/2022] Open
Abstract
The aging brain seems to be characterized by neuronal loss leading to cognitive decline and progressively worsening symptoms related to neurodegeneration. Also, pro-inflammatory states, if prolonged, may increase neuronal vulnerability via excessive activation of microglia and their pro-inflammatory by-products, which is seen as individuals increase in age. Consequently, microglial activity is tightly regulated by neuron-microglia communications. The endocannabinoid system (ECS) is emerging as a regulator of microglia and the neuronal-microglia communication system. Recently, it has been demonstrated that cannabinoid 1 (CB1) receptor signaling on GABAergic interneurons plays a crucial role in regulating microglial activity. Interestingly, if endocannabinoid signaling on GABAergic neurons are disturbed, the phenotypes mimic central nervous system insult models by activating microglia and leading to accelerated brain aging. Investigating the endocannabinoid receptors, ligands, and genetic deletions yields the potential to understand the communication system and mechanism by which the ECS regulates glial cells and aspects of aging. While there remains much to discover with the ECS, the information gathered and identified already could lead to the development of cell-specific therapeutic interventions that help in reducing the effects of age-related pro-inflammatory states and neurodegeneration.
Collapse
Affiliation(s)
- Jorge Carrera
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Jensen Tomberlin
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States.,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - John Kurtz
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Eda Karakaya
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | | | - Onder Albayram
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States.,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States.,Ralph H. Johnson VA Medical Center, Charleston, SC, United States
| |
Collapse
|
28
|
Tampio J, Huttunen J, Montaser A, Huttunen KM. Targeting of Perforin Inhibitor into the Brain Parenchyma Via a Prodrug Approach Can Decrease Oxidative Stress and Neuroinflammation and Improve Cell Survival. Mol Neurobiol 2020; 57:4563-4577. [PMID: 32754897 PMCID: PMC7515946 DOI: 10.1007/s12035-020-02045-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/28/2020] [Indexed: 12/31/2022]
Abstract
The cytolytic protein perforin has a crucial role in infections and tumor surveillance. Recently, it has also been associated with many brain diseases, such as neurodegenerative diseases and stroke. Therefore, inhibitors of perforin have attracted interest as novel drug candidates. We have previously reported that converting a perforin inhibitor into an L-type amino acid transporter 1 (LAT1)-utilizing prodrug can improve the compound's brain drug delivery not only across the blood-brain barrier (BBB) but also into the brain parenchymal cells: neurons, astrocytes, and microglia. The present study evaluated whether the increased uptake into mouse primary cortical astrocytes and subsequently improvements in the cellular bioavailability of this brain-targeted perforin inhibitor prodrug could enhance its pharmacological effects, such as inhibition of production of caspase-3/-7, lipid peroxidation products and prostaglandin E2 (PGE2) in the lipopolysaccharide (LPS)-induced neuroinflammation mouse model. It was demonstrated that increased brain and cellular drug delivery could improve the ability of perforin inhibitors to elicit their pharmacological effects in the brain at nano- to picomolar levels. Furthermore, the prodrug displayed multifunctional properties since it also inhibited the activity of several key enzymes related to Alzheimer's disease (AD), such as the β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), acetylcholinesterase (AChE), and most probably also cyclooxygenases (COX) at micromolar concentrations. Therefore, this prodrug is a potential drug candidate for preventing Aβ-accumulation and ACh-depletion in addition to combatting neuroinflammation, oxidative stress, and neural apoptosis within the brain. Graphical abstract.
Collapse
Affiliation(s)
- Janne Tampio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Ahmed Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
29
|
Val-Laillet D, Kanzari A, Guérin S, Randuineau G, Coquery N. A maternal Western diet during gestation and lactation modifies offspring's microglial cell density and morphology in the hippocampus and prefrontal cortex in Yucatan minipigs. Neurosci Lett 2020; 739:135395. [PMID: 32950568 DOI: 10.1016/j.neulet.2020.135395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
Changes in microglial development and morphology can be induced by inflammatory conditions and associated with eating or mood disorders, such as hyperphagia or depression. In a previous paper in the minipig model, we showed that maternal Western diet during gestation and lactation decreased hippocampus neurogenesis and food-rewarded cognitive abilities in the progeny. Whether these alterations are concomitant with a central inflammatory process in brain structures involved in learning and memory (hippocampus, HPC), cognitive (prefrontal cortex, PFC), or hedonic (orbitofrontal cortex, OFC) control of food intake is still unknown. In the present study, Yucatan minipigs (Sus scrofa) sows were exposed to two different diets during gestation and lactation (standard, SD N = 7 vs. Western diet, WD N = 9). Iba1 is a calcium-binding protein specifically expressed in microglia in the brain, which plays an important role in the regulation of the microglia function. Iba1 expression was examined by immunohistochemical analyses in the PFC, OFC and HPC of piglets. The density of microglial cells, as well as their morphology, were assessed in order to have an indirect insight of microglial cell activation state possibly in relationship with neuroinflammation. The density of Iba1-positive cells was higher in the PFC but not in the HPC of WD compared to SD piglets (p < 0.001). In the HPC, anterior and dorsolateral PFC, WD piglets had more unipolar cells, contrary to SD that had more multipolar cells (P < 0.0001). Opposite effects were observed in the OFC, with SD presenting more unipolar (P < 0.001) microglial cells compared to WD. We showed here that maternal diet during pregnancy and lactation had significant effects on morphological changes of microglial cells in the offspring, and that these effects differed between the HPC and PFC, suggesting different response mechanisms to the early nutritional environment.
Collapse
Affiliation(s)
- David Val-Laillet
- INRAE, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, St Gilles, France.
| | - Ameni Kanzari
- INRAE, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, St Gilles, France; Université de Tunis El Manar, Faculté des Sciences de Tunis, UR/11ES09 Laboratory of Functional Neurophysiology and Pathology, Tunis, Tunisia
| | - Sylvie Guérin
- INRAE, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, St Gilles, France
| | - Gwénaëlle Randuineau
- INRAE, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, St Gilles, France
| | - Nicolas Coquery
- INRAE, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, St Gilles, France
| |
Collapse
|
30
|
Tan YL, Yuan Y, Tian L. Microglial regional heterogeneity and its role in the brain. Mol Psychiatry 2020; 25:351-367. [PMID: 31772305 PMCID: PMC6974435 DOI: 10.1038/s41380-019-0609-8] [Citation(s) in RCA: 317] [Impact Index Per Article: 63.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023]
Abstract
Microglia have been recently shown to manifest a very interesting phenotypical heterogeneity across different regions in the mammalian central nervous system (CNS). However, the underlying mechanism and functional meaning of this phenomenon are currently unclear. Baseline diversities of adult microglia in their cell number, cellular and subcellular structures, molecular signature as well as relevant functions have been discovered. But recent transcriptomic studies using bulk RNAseq and single-cell RNAseq have produced conflicting results on region-specific signatures of microglia. It is highly speculative whether such spatial heterogeneity contributes to varying sensitivities of individual microglia to the same physiological and pathological signals in different CNS regions, and hence underlie their functional relevance for CNS disease development. This review aims to thoroughly summarize up-to-date knowledge on this specific topic and provide some insights on the potential underlying mechanisms, starting from microgliogenesis. Understanding regional heterogeneity of microglia in the context of their diverse neighboring neurons and other glia may provide an important clue for future development of innovative therapies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yun-Long Tan
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Yi Yuan
- Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Li Tian
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China.
- Institute of Biomedicine and Translational Medicine, Department of Physiology, Faculty of Medicine, University of Tartu, Tartu, Estonia.
| |
Collapse
|
31
|
Kikuchi T, Gonzalez-Soriano J, Kastanauskaite A, Benavides-Piccione R, Merchan-Perez A, DeFelipe J, Blazquez-Llorca L. Volume Electron Microscopy Study of the Relationship Between Synapses and Astrocytes in the Developing Rat Somatosensory Cortex. Cereb Cortex 2020; 30:3800-3819. [PMID: 31989178 PMCID: PMC7233003 DOI: 10.1093/cercor/bhz343] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
In recent years, numerous studies have shown that astrocytes play an important role in neuronal processing of information. One of the most interesting findings is the existence of bidirectional interactions between neurons and astrocytes at synapses, which has given rise to the concept of “tripartite synapses” from a functional point of view. We used focused ion beam milling and scanning electron microscopy (FIB/SEM) to examine in 3D the relationship of synapses with astrocytes that were previously labeled by intracellular injections in the rat somatosensory cortex. We observed that a large number of synapses (32%) had no contact with astrocytic processes. The remaining synapses (68%) were in contact with astrocytic processes, either at the level of the synaptic cleft (44%) or with the pre- and/or post-synaptic elements (24%). Regarding synaptic morphology, larger synapses with more complex shapes were most frequently found within the population that had the synaptic cleft in contact with astrocytic processes. Furthermore, we observed that although synapses were randomly distributed in space, synapses that were free of astrocytic processes tended to form clusters. Overall, at least in the developing rat neocortex, the concept of tripartite synapse only seems to be applicable to a subset of synapses.
Collapse
Affiliation(s)
- Toko Kikuchi
- Center for Biosciences and Informatics, School of Fundamental Science and Technology, Graduate School of Science and Technology, Keio University, 223-8522 Kanagawa, Japan.,Department of Fundamental Neuroscience, University of Lausanne, 1015 Lausanne, Switzerland
| | - Juncal Gonzalez-Soriano
- Departamento de Anatomía, Facultad de Veterinaria, Universidad Complutense, 28040 Madrid, Spain
| | - Asta Kastanauskaite
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Ruth Benavides-Piccione
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain.,Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, 28002 Madrid, Spain
| | - Angel Merchan-Perez
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain.,Departamento de Arquitectura y Tecnología de Sistemas Informáticos, Escuela Técnica Superior de Ingenieros Informáticos, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain.,Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, 28002 Madrid, Spain
| | - Lidia Blazquez-Llorca
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Madrid, Spain.,Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain
| |
Collapse
|
32
|
Astrocyte-Targeted Transporter-Utilizing Derivatives of Ferulic Acid Can Have Multifunctional Effects Ameliorating Inflammation and Oxidative Stress in the Brain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3528148. [PMID: 31814871 PMCID: PMC6877910 DOI: 10.1155/2019/3528148] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/19/2019] [Accepted: 10/03/2019] [Indexed: 12/27/2022]
Abstract
Ferulic acid (FA) is a natural phenolic antioxidant, which can exert also several other beneficial effects to combat neuroinflammation and neurodegenerative diseases, such as Alzheimer's disease. One of these properties is the inhibition of several enzymes and factors, such as β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), cyclooxygenases (COXs), lipoxygenases (LOXs), mammalian (or mechanistic) target for rapamycin (mTOR), and transcription factor NF-κB. We have previously synthesized three L-type amino acid transporter 1- (LAT1-) utilizing FA-derivatives with the aim to develop brain-targeted prodrugs of FA. In the present study, the cellular uptake and bioavailability of these FA-derivatives were evaluated in mouse primary astrocytic cell cultures together with their inhibitory effects towards BACE1, COX/LOX, mTOR, NF-κB, acetylcholinesterase (AChE), and oxidative stress. According to the results, all three FA-derivatives were taken up 200–600 times more effectively at 10 μM concentration into the astrocytes than FA, with one derivative having a high intracellular bioavailability (Kp,uu), particularly at low concentrations. Moreover, all of the derivatives were able to inhibit BACE1, COX/LOX, AChE, and oxidative stress measured as decreased cellular lipid peroxidation. Furthermore, one of the derivatives modified the total mTOR amount. Therefore, these derivatives have the potential to act as multifunctional compounds preventing β-amyloid accumulation as well as combating inflammation and reducing oxidative stress in the brain. Thus, this study shows that converting a parent drug into a transporter-utilizing derivative not only may increase its brain and cellular uptake, and bioavailability but can also broaden the spectrum of pharmacological effects elicited by the derivative.
Collapse
|
33
|
Attili SM, Silva MFM, Nguyen TV, Ascoli GA. Cell numbers, distribution, shape, and regional variation throughout the murine hippocampal formation from the adult brain Allen Reference Atlas. Brain Struct Funct 2019; 224:2883-2897. [PMID: 31444616 PMCID: PMC6778719 DOI: 10.1007/s00429-019-01940-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/13/2019] [Indexed: 01/07/2023]
Abstract
Quantifying the distribution of cells in every brain region is fundamental to attaining a comprehensive census of distinct neuronal and glial types. Until recently, estimating neuron numbers involved time-consuming procedures that were practically limited to stereological sampling. Progress in open-source image recognition software, growth in computing power, and unprecedented neuroinformatics developments now offer the potentially paradigm-shifting alternative of comprehensive cell-by-cell analysis in an entire brain region. The Allen Brain Atlas provides free digital access to complete series of raw Nissl-stained histological section images along with regional delineations. Automated cell segmentation of these data enables reliable and reproducible high-throughput quantification of regional variations in cell count, density, size, and shape at whole-system scale. While this strategy is directly applicable to any regions of the mouse brain, we first deploy it here on the closed-loop circuit of the hippocampal formation: the medial and lateral entorhinal cortices; dentate gyrus (DG); areas Cornu Ammonis 3 (CA3), CA2, and CA1; and dorsal and ventral subiculum. Using two independent image processing pipelines and the adult mouse reference atlas, we report the first cellular-level soma segmentation in every sub-region and non-principal layer of the left hippocampal formation through the full rostral-caudal extent. It is important to note that our techniques excluded the layers with the largest number of cells, DG granular and CA pyramidal, due to dense packing. The numerical estimates for the remaining layers are corroborated by traditional stereological sampling on a data subset and well match sparse published reports.
Collapse
Affiliation(s)
- Sarojini M Attili
- Center for Neural Informatics Structures and Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Marcos F M Silva
- Center for Neural Informatics Structures and Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Thuy-Vi Nguyen
- Center for Neural Informatics Structures and Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
- Duke University, Durham, NC, USA
| | - Giorgio A Ascoli
- Center for Neural Informatics Structures and Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA.
| |
Collapse
|
34
|
Jacobs AJ, Castillo‐Ruiz A, Cisternas CD, Forger NG. Microglial Depletion Causes Region‐Specific Changes to Developmental Neuronal Cell Death in the Mouse Brain. Dev Neurobiol 2019; 79:769-779. [DOI: 10.1002/dneu.22706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/17/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Andrew J. Jacobs
- Neuroscience Institute Georgia State University P.O. Box 5030 Atlanta Georgia30302‐5030
| | | | - Carla D. Cisternas
- Neuroscience Institute Georgia State University P.O. Box 5030 Atlanta Georgia30302‐5030
| | - Nancy G. Forger
- Neuroscience Institute Georgia State University P.O. Box 5030 Atlanta Georgia30302‐5030
| |
Collapse
|
35
|
O'Koren EG, Yu C, Klingeborn M, Wong AYW, Prigge CL, Mathew R, Kalnitsky J, Msallam RA, Silvin A, Kay JN, Bowes Rickman C, Arshavsky VY, Ginhoux F, Merad M, Saban DR. Microglial Function Is Distinct in Different Anatomical Locations during Retinal Homeostasis and Degeneration. Immunity 2019; 50:723-737.e7. [PMID: 30850344 DOI: 10.1016/j.immuni.2019.02.007] [Citation(s) in RCA: 266] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/03/2018] [Accepted: 02/12/2019] [Indexed: 01/09/2023]
Abstract
Microglia from different nervous system regions are molecularly and anatomically distinct, but whether they also have different functions is unknown. We combined lineage tracing, single-cell transcriptomics, and electrophysiology of the mouse retina and showed that adult retinal microglia shared a common developmental lineage and were long-lived but resided in two distinct niches. Microglia in these niches differed in their interleukin-34 dependency and functional contribution to visual-information processing. During certain retinal-degeneration models, microglia from both pools relocated to the subretinal space, an inducible disease-associated niche that was poorly accessible to monocyte-derived cells. This microglial transition involved transcriptional reprogramming of microglia, characterized by reduced expression of homeostatic checkpoint genes and upregulation of injury-responsive genes. This transition was associated with protection of the retinal pigmented epithelium from damage caused by disease. Together, our data demonstrate that microglial function varies by retinal niche, thereby shedding light on the significance of microglia heterogeneity.
Collapse
Affiliation(s)
- Emily G O'Koren
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA
| | - Chen Yu
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA
| | | | - Alicia Y W Wong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Cameron L Prigge
- Department of Neurobiology, Duke University, Durham, NC 27710, USA
| | - Rose Mathew
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA
| | - Joan Kalnitsky
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA
| | - Rasha A Msallam
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Aymeric Silvin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Jeremy N Kay
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; Department of Neurobiology, Duke University, Durham, NC 27710, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; Department of Pharmacology, Duke University, Durham, NC 27710, USA
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Miriam Merad
- Precision Immunology Institute and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniel R Saban
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
36
|
Izquierdo P, Attwell D, Madry C. Ion Channels and Receptors as Determinants of Microglial Function. Trends Neurosci 2019; 42:278-292. [PMID: 30678990 DOI: 10.1016/j.tins.2018.12.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022]
Abstract
Microglia provide immune surveillance of the CNS. They display diverse behaviors, including nondirectional and directed motility of their processes, phagocytosis of targets such as dying neurons or superfluous synapses, and generation of reactive oxygen species (ROS) and cytokines. Many of these functions are mediated by ion channels and cell surface receptors, the expression of which varies with the many morphological and functional states that microglial cells can adopt. Recent progress in understanding microglial function has been facilitated by applying classical cell physiological techniques in situ, such as patch-clamping and live imaging, and cell-specific transcriptomic analyses. Here, we review the contribution of microglial ion channels and receptors to microglial and brain function.
Collapse
Affiliation(s)
- Pablo Izquierdo
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower St, London, WC1E 6BT, UK
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower St, London, WC1E 6BT, UK.
| | - Christian Madry
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117 Berlin, Germany.
| |
Collapse
|
37
|
Iida T, Tanaka S, Okabe S. Spatial impact of microglial distribution on dynamics of dendritic spines. Eur J Neurosci 2019; 49:1400-1417. [DOI: 10.1111/ejn.14325] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/10/2018] [Accepted: 12/20/2018] [Indexed: 01/31/2023]
Affiliation(s)
- Tadatsune Iida
- Department of Cellular Neurobiology Graduate School of Medicine The University of Tokyo Tokyo Japan
| | - Shinji Tanaka
- Department of Cellular Neurobiology Graduate School of Medicine The University of Tokyo Tokyo Japan
| | - Shigeo Okabe
- Department of Cellular Neurobiology Graduate School of Medicine The University of Tokyo Tokyo Japan
| |
Collapse
|
38
|
Rashidiani-Rashidabadi A, Heidari MH, Sajadi E, Hejazi F, Fathabady FF, Sadeghi Y, Aliaghaei A, Raoofi A, Abdollahifar MA, Farahni RM. Sciatic nerve injury alters the spatial arrangement of neurons and glial cells in the anterior horn of the spinal cord. Neural Regen Res 2019; 14:1833-1840. [PMID: 31169202 PMCID: PMC6585558 DOI: 10.4103/1673-5374.257539] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The spatial arrangement of the cell is important and considered as underlying mechanism for mathematical modeling of cell to cell interaction. The ability of cells to take on the characteristics of other cells in an organism, it is important to understand the dynamical behavior of the cells. This method implements experimental parameters of the cell-cell interaction into the mathematical simulation of cell arrangement. The purpose of this research was to explore the three-dimensional spatial distribution of anterior horn cells in the rat spinal cord to examine differences after sciatic nerve injury. Sixteen Sprague-Dawley male rats were assigned to control and axotomy groups. Twelve weeks after surgery, the anterior horn was removed for first- and second-order stereological studies. Second-order stereological techniques were applied to estimate the pair correlation and cross-correlation functions using a dipole probe superimposed onto the spinal cord sections. The findings revealed 7% and 36% reductions in the mean volume and total number of motoneurons, respectively, and a 25% increase in the neuroglial cell number in the axotomized rats compared to the control rats. In contrast, the anterior horn volume remained unchanged. The results also indicated a broader gap in the pair correlation curve for the motoneurons and neuroglial cells in the axotomized rats compared to the control rats. This finding shows a negative correlation for the distribution of motoneurons and neuroglial cells in the axotomized rats. The cross-correlation curve shows a negative correlation between the motoneurons and neuroglial cells in the axotomized rats. These findings suggest that cellular structural and functional changes after sciatic nerve injury lead to the alterations in the spatial arrangement of motoneurons and neuroglial cells, finally affecting the normal function of the central nervous system. The experimental protocol was reviewed and approved by the Animal Ethics Committee of Shahid Beheshti University of Medical Sciences (approval No. IR.SBMU.MSP.REC1395.375) on October 17, 2016.
Collapse
Affiliation(s)
- Ali Rashidiani-Rashidabadi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassan Heidari
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ensieh Sajadi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Hejazi
- Department of Polymer Engineering and Color Technology, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Fatemeh Fadaei Fathabady
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yousef Sadeghi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Raoofi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Mastery Farahni
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
A comparison of hippocampal microglial responses in aged and young rodents following dependent and non-dependent binge drinking. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:305-343. [PMID: 31733666 PMCID: PMC9875180 DOI: 10.1016/bs.irn.2019.10.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Alcoholism is a highly visible and prevalent issue in the United States. Although binge-drinking is assumed to be a college-age problem, older adults (ages 65+) consume binge amounts of alcohol and have alcohol use disorders (AUDs). Moreover, individuals with alcohol dependence in their youth often continue to drink as they age. As such, this study tested the hypothesis that the effects of alcohol on hippocampal microglia are exacerbated in aged versus younger rodents in two AUD models. Briefly, adult (2-3 months) and aged (15+ months) Sprague-Dawley rats were administered alcohol or control diet using the Majchrowicz model to study alcohol-induced neurodegeneration. To study the effects of non-dependent binge consumption on microglia, adolescent (6-8 weeks) and aged (18+ months) C57/BL6N were subjected to the Drinking in the Dark paradigm. Microglia number and densitometry were assessed using immunohistochemistry. Hippocampal subregional and model/species-specific effects of alcohol were observed, but overall, aging did not appear to increase the alcohol-induced microglia reactivity as measured by Iba-1 densitometry. However, analysis of microglial counts revealed a significant decrease in the number microglia cells in both the alcohol-induced neurodegeneration and DID model across age groups. In the dentate gyrus, the loss of microglia was exacerbated by aging, particularly in mice after DID, non-dependent model. Using qRT-PCR, the persistence of alcohol and aging effects was assessed following the DID model. Allograft Inflammatory Factor 1 mRNA was increased in both young and aged mice by alcohol exposure; however, only in the aged mice did the alcohol effect persist. Overall, these data imply that the microglial response to alcohol is complex with evidence of depressed numbers of microglia but also increased reactivity with advanced age.
Collapse
|
40
|
Keller D, Erö C, Markram H. Cell Densities in the Mouse Brain: A Systematic Review. Front Neuroanat 2018; 12:83. [PMID: 30405363 PMCID: PMC6205984 DOI: 10.3389/fnana.2018.00083] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/20/2018] [Indexed: 11/29/2022] Open
Abstract
The mouse brain is the most extensively studied brain of all species. We performed an exhaustive review of the literature to establish our current state of knowledge on cell numbers in mouse brain regions, arguably the most fundamental property to measure when attempting to understand a brain. The synthesized information, collected in one place, can be used by both theorists and experimentalists. Although for commonly-studied regions cell densities could be obtained for principal cell types, overall we know very little about how many cells are present in most brain regions and even less about cell-type specific densities. There is also substantial variation in cell density values obtained from different sources. This suggests that we need a new approach to obtain cell density datasets for the mouse brain.
Collapse
Affiliation(s)
- Daniel Keller
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | | | | |
Collapse
|
41
|
Dominguez S, Flores-Montoya MG, Sobin C. Early chronic exposure to low-level lead alters total hippocampal microglia in pre-adolescent mice. Toxicol Lett 2018; 302:75-82. [PMID: 30352268 DOI: 10.1016/j.toxlet.2018.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/28/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022]
Abstract
Developmental lead (Pb) exposure alters brain function through mechanisms that are not yet understood. A previous study showed that early lead exposure reduced microglia number in the dentate gyrus region of the hippocampus. Given the critical role of microglia in brain development, it is important to determine whether these differences are unique to the dentate gyrus, or occur throughout the hippocampus. Unbiased stereology was used to quantify microglia mean cell body number in total hippocampus, and compare the proportion of microglia in the ventral vs. dorsal regions. Total hippocampal volume was also measured and compared. The study included brain tissue from 30 pre-adolescent C57BL/6 J mice, exposed to 30 ppm Pb acetate (n = 10, mean BLL 3.4 μg/dL at sacrifice), 330 ppm Pb acetate (n = 10, mean BLL 14.1 μg/dL at sacrifice), or 0 ppm Pb acetate (n = 10, negative controls). In lead exposed animals, microglia mean cell body number was reduced in total hippocampus; total hippocampal volume was reduced. Importantly, effects in low- and high-dose exposure groups did not differ. Contrary to study hypotheses, the distribution of hippocampal microglia in the ventral vs. dorsal hippocampal regions did not differ. Overall, lowest and higher levels of lead exposure during development had strikingly similar disruptive effects in the neuroimmune system. Studies are needed to determine the immune and other mechanisms responsible for these effects. Future studies would benefit from larger samples to determine whether in fact there is a group by sex interaction driving the effects of early lead exposure on microglia.
Collapse
Affiliation(s)
- Salvador Dominguez
- Dept of Public Health Sciences, University of Texas, El Paso, TX, United States; Border Biomedical Research Center, Toxicology Core, University of Texas, El Paso, TX, United States
| | - Mayra Gisel Flores-Montoya
- Border Biomedical Research Center, Toxicology Core, University of Texas, El Paso, TX, United States; Dept of Psychology, University of Texas, El Paso, TX, United States
| | - Christina Sobin
- Dept of Public Health Sciences, University of Texas, El Paso, TX, United States; Border Biomedical Research Center, Toxicology Core, University of Texas, El Paso, TX, United States; Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, United States.
| |
Collapse
|
42
|
Yang EJ, Mahmood U, Kim H, Choi M, Choi Y, Lee JP, Cho JY, Hyun JW, Kim YS, Chang MJ, Kim HS. Phloroglucinol ameliorates cognitive impairments by reducing the amyloid β peptide burden and pro-inflammatory cytokines in the hippocampus of 5XFAD mice. Free Radic Biol Med 2018; 126:221-234. [PMID: 30118828 DOI: 10.1016/j.freeradbiomed.2018.08.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 01/29/2023]
Abstract
Among the various causative factors involved in the pathogenesis of Alzheimer's disease (AD), oxidative stress has emerged as an important factor. Phloroglucinol is a polyphenol component of phlorotannin, which is found at sufficient levels in Ecklonia cava (E. cava). Phloroglucinol has been reported to exert antioxidant activities in various tissues. Previously, we reported that the stereotaxic injection of phloroglucinol regulated synaptic plasticity in an AD mouse model. In this study, we aimed to investigate the effects of oral administration of phloroglucinol in AD. The oral administration of phloroglucinol for 2 months attenuated the impairments in cognitive function observed in 6-month-old 5X familial AD (5XFAD) mice, as assessed with the T-maze and Y-maze tests. The administration of phloroglucinol for 2 months in 5XFAD mice caused a reduction in the number of amyloid plaques and in the protein level of BACE1, a major amyloid precursor protein cleavage enzyme, together with γ-secretase. Phloroglucinol also restored the reduction in dendritic spine density and the number of mature spines in the hippocampi of 5XFAD mice. In addition, phloroglucinol-administered 5XFAD mice displayed lower protein levels of GFAP and Iba-1 and mRNA levels of TNF-α and IL-6 compared with vehicle-administered 5XFAD mice. These results demonstrated that phloroglucinol alleviated the neuropathological features and behavioral phenotypes in the 5XFAD mouse model. Taken together, our results suggest that phloroglucinol has therapeutic potential for AD treatment.
Collapse
Affiliation(s)
- Eun-Jeong Yang
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Usman Mahmood
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Hyunju Kim
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Moonseok Choi
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Yunjung Choi
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Jean-Pyo Lee
- Department of Physiology, Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
| | - Joo-Youn Cho
- Department of Clinical Pharmacology & Therapeutics, College of Medicine, Seoul National University and Hospital, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Yong Sik Kim
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Moon-Jeong Chang
- Department of Foods and Nutrition, College of Natural Science, Kookmin University, Seoul 02707, Republic of Korea.
| | - Hye-Sun Kim
- Department of Pharmacology, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea; Seoul National University Bundang Hospital, Seongnam, Sungnam, Bundang-Gu 13620, Republic of Korea; Neuroscience Research Institute, Seoul National University, College of Medicine, 103 Daehakro, Jongro-gu, Seoul 03080, Republic of Korea.
| |
Collapse
|
43
|
Filipello F, Morini R, Corradini I, Zerbi V, Canzi A, Michalski B, Erreni M, Markicevic M, Starvaggi-Cucuzza C, Otero K, Piccio L, Cignarella F, Perrucci F, Tamborini M, Genua M, Rajendran L, Menna E, Vetrano S, Fahnestock M, Paolicelli RC, Matteoli M. The Microglial Innate Immune Receptor TREM2 Is Required for Synapse Elimination and Normal Brain Connectivity. Immunity 2018; 48:979-991.e8. [PMID: 29752066 DOI: 10.1016/j.immuni.2018.04.016] [Citation(s) in RCA: 429] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/19/2018] [Accepted: 04/13/2018] [Indexed: 02/07/2023]
Abstract
The triggering receptor expressed on myeloid cells 2 (TREM2) is a microglial innate immune receptor associated with a lethal form of early, progressive dementia, Nasu-Hakola disease, and with an increased risk of Alzheimer's disease. Microglial defects in phagocytosis of toxic aggregates or apoptotic membranes were proposed to be at the origin of the pathological processes in the presence of Trem2 inactivating mutations. Here, we show that TREM2 is essential for microglia-mediated synaptic refinement during the early stages of brain development. The absence of Trem2 resulted in impaired synapse elimination, accompanied by enhanced excitatory neurotransmission and reduced long-range functional connectivity. Trem2-/- mice displayed repetitive behavior and altered sociability. TREM2 protein levels were also negatively correlated with the severity of symptoms in humans affected by autism. These data unveil the role of TREM2 in neuronal circuit sculpting and provide the evidence for the receptor's involvement in neurodevelopmental diseases.
Collapse
Affiliation(s)
- Fabia Filipello
- Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini, 20090 Pieve Emanuele - Milan, Italy
| | - Raffaella Morini
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano - Milan, Italy
| | - Irene Corradini
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano - Milan, Italy; IN-CNR, 20129 Milano, Italy
| | - Valerio Zerbi
- Neural Control of Movement Lab, HEST, ETH Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Alice Canzi
- Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini, 20090 Pieve Emanuele - Milan, Italy
| | - Bernadeta Michalski
- Department of Psychiatry & Behavioural Neurosciences, HSC-4N80, McMaster University, Hamilton, ON, Canada
| | - Marco Erreni
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano - Milan, Italy
| | - Marija Markicevic
- Neural Control of Movement Lab, HEST, ETH Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Chiara Starvaggi-Cucuzza
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano - Milan, Italy
| | - Karel Otero
- Department of Neuroimmunology, Acute Neurology and Pain, Biogen Inc., 115 Broadway, Cambridge, MA, USA
| | - Laura Piccio
- Department of Neurology, Washington University, St. Louis, MO, USA
| | | | - Fabio Perrucci
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano - Milan, Italy
| | - Matteo Tamborini
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano - Milan, Italy
| | - Marco Genua
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano - Milan, Italy
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, IREM, University of Zurich, Schlieren, Switzerland
| | - Elisabetta Menna
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano - Milan, Italy; IN-CNR, 20129 Milano, Italy
| | - Stefania Vetrano
- Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini, 20090 Pieve Emanuele - Milan, Italy
| | - Margaret Fahnestock
- Department of Psychiatry & Behavioural Neurosciences, HSC-4N80, McMaster University, Hamilton, ON, Canada
| | - Rosa Chiara Paolicelli
- Systems and Cell Biology of Neurodegeneration, IREM, University of Zurich, Schlieren, Switzerland
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano - Milan, Italy; IN-CNR, 20129 Milano, Italy.
| |
Collapse
|
44
|
DeWalt GJ, Mahajan B, Foster AR, Thompson LDE, Marttini AA, Schmidt EV, Mansuri S, D'Souza D, Patel SB, Tenenbaum M, Brandao-Viruet KI, Thompson D, Duong B, Smith DH, Blute TA, Eldred WD. Region-specific alterations in astrocyte and microglia morphology following exposure to blasts in the mouse hippocampus. Neurosci Lett 2017; 664:160-166. [PMID: 29133177 DOI: 10.1016/j.neulet.2017.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/28/2022]
Abstract
Traumatic brain injury (TBI) is a serious public health concern, especially injuries from repetitive insults. The main objective of this study was to immunocytochemically examine morphological alterations in astrocytes and microglia in the hippocampus 48h following a single blast versus multiple blasts in adult C57BL/6 mice. The effects of ketamine and xylazine (KX), two common anesthetic agents used in TBI research, were also evaluated due to the confounding effect of anesthetics on injury outcome. Results showed a significant increase in hypertrophic microglia that was limited to the outer molecular layer of the dentate gyrus, but only in the absence of KX. Although the presence or absence of KX had no effect on astrocytes following a single blast, a significant decrease in astrocytic immunoreactivity was observed in the stratum lacunosum moleculare following multiple blasts in the absence of KX. The morphological changes in astrocytes and microglia reported in this study reveal region-specific differences in the absence of KX that could have significant implications for our interpretation of glial alterations in animal models of injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sara Mansuri
- Boston University, Department of Biology, United States
| | | | - Shama B Patel
- Boston University, Department of Biology, United States
| | | | | | | | - Bryan Duong
- Boston University, Department of Biology, United States
| | | | - Todd A Blute
- Boston University, Department of Biology, United States
| | | |
Collapse
|
45
|
Maurer SV, Williams CL. The Cholinergic System Modulates Memory and Hippocampal Plasticity via Its Interactions with Non-Neuronal Cells. Front Immunol 2017; 8:1489. [PMID: 29167670 PMCID: PMC5682336 DOI: 10.3389/fimmu.2017.01489] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/23/2017] [Indexed: 01/12/2023] Open
Abstract
Degeneration of central cholinergic neurons impairs memory, and enhancement of cholinergic synapses improves cognitive processes. Cholinergic signaling is also anti-inflammatory, and neuroinflammation is increasingly linked to adverse memory, especially in Alzheimer's disease. Much of the evidence surrounding cholinergic impacts on the neuroimmune system focuses on the α7 nicotinic acetylcholine (ACh) receptor, as stimulation of this receptor prevents many of the effects of immune activation. Microglia and astrocytes both express this receptor, so it is possible that some cholinergic effects may be via these non-neuronal cells. Though the presence of microglia is required for memory, overactivated microglia due to an immune challenge overproduce inflammatory cytokines, which is adverse for memory. Blocking these exaggerated effects, specifically by decreasing the release of tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), and interleukin 6 (IL-6), has been shown to prevent inflammation-induced memory impairment. While there is considerable evidence that cholinergic signaling improves memory, fewer studies have linked the "cholinergic anti-inflammatory pathway" to memory processes. This review will summarize the current understanding of the cholinergic anti-inflammatory pathway as it relates to memory and will argue that one mechanism by which the cholinergic system modulates hippocampal memory processes is its influence on neuroimmune function via the α7 nicotinic ACh receptor.
Collapse
Affiliation(s)
- Sara V. Maurer
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| | - Christina L. Williams
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| |
Collapse
|
46
|
De Luca SN, Ziko I, Dhuna K, Sominsky L, Tolcos M, Stokes L, Spencer SJ. Neonatal overfeeding by small-litter rearing sensitises hippocampal microglial responses to immune challenge: Reversal with neonatal repeated injections of saline or minocycline. J Neuroendocrinol 2017; 29. [PMID: 28983991 DOI: 10.1111/jne.12540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/22/2017] [Accepted: 10/01/2017] [Indexed: 01/08/2023]
Abstract
The early-life period is extremely vulnerable to programming effects from the environment, many of which persist into adulthood. We have previously demonstrated that adult rats overfed as neonates have hypothalamic microglia that are hyper-responsive to an immune challenge, as well as hippocampal microglia that respond less efficiently to learning. We therefore hypothesised that neonatal overfeeding would alter the ability of hippocampal microglia to respond to an immune challenge with lipopolysaccharide (LPS) and that concomitant minocycline, a tetracycline antibiotic that suppresses microglial activity, could restore these responses. We induced neonatal overfeeding by manipulating the litter sizes in which Wistar rat pups were raised, so the pups were suckled in litters of four (neonatally overfed) or 12 (control-fed). We then examined the hippocampal microglial profiles 24 hour after an immune challenge with LPS and found that the neonatally overfed rats had dramatically increased microglial numbers in the hippocampus after immune challenge compared to control-fed rats. Attempts to reverse these effects with minocycline revealed repeated that neonatal injections, whether with minocycline or with saline, markedly suppressed microglial number and density throughout the hippocampus and abolished the difference between the groups in their responses to LPS. These data suggest that neonatal overfeeding not only can have lasting effects on hippocampal immune responses, but also that neonatal exposure to a protocol of repeated injections, irrespective of treatment, has a pronounced long-term impact, highlighting the importance of considering these effects when interpreting experimental data.
Collapse
Affiliation(s)
- S N De Luca
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - I Ziko
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - K Dhuna
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - L Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - M Tolcos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - L Stokes
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, UK
| | - S J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
47
|
Gzielo K, Kielbinski M, Ploszaj J, Janeczko K, Gazdzinski SP, Setkowicz Z. Long-Term Consumption of High-Fat Diet in Rats: Effects on Microglial and Astrocytic Morphology and Neuronal Nitric Oxide Synthase Expression. Cell Mol Neurobiol 2017; 37:783-789. [PMID: 27541371 PMCID: PMC5435787 DOI: 10.1007/s10571-016-0417-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/10/2016] [Indexed: 12/30/2022]
Abstract
Obesity in humans is associated with cognitive decline and elevated risk of neurodegenerative diseases of old age. Variations of high-fat diet are often used to model these effects in animal studies. However, we previously reported improvements in markers of memory and learning, as well as larger hippocampi and higher metabolite concentrations in Wistar rats fed high-fat, high-carbohydrate diet (HFCD, 60 % energy from fat, 28 % from carbohydrates) for 1 year; this diet leads to mild ketonemia (Setkowicz et al. in PLoS One 10:e0139987, 2015). In the present study, we follow up on this cohort to assess glial morphology and expression of markers related to gliosis. Twenty-five male Wistar rats were kept on HFCD and twenty-five on normal chow. At 12 months of age, the animals were sacrificed and processed for immunohistochemical staining for astrocytic (glial fibrillary acidic protein), microglial (Iba1), and neuronal (neuronal nitric oxide synthetase, nNOS) markers in the hippocampus. We have found changes in immunopositive area fraction and cellular complexity, as studied by a simplified Sholl procedure. To our knowledge, this study is the first to apply this methodology to the study of glial cells in HFCD animals. GFAP and Iba1 immunoreactive area fraction in the hippocampi of HFCD-fed rats were decreased, while the mean number of intersections (an indirect measure of cell complexity) was decreased in GFAP-positive astrocytes, but not in Iba1-expressing microglia. At the same time, nNOS expression was lowered after HFCD in both the cortex and the hippocampus.
Collapse
Affiliation(s)
- Kinga Gzielo
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland.
| | - Michal Kielbinski
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Jakub Ploszaj
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Krzysztof Janeczko
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Stefan P Gazdzinski
- Military Institute of Aviation Medicine, Krasinskiego 54, 01-755, Warsaw, Poland
| | - Zuzanna Setkowicz
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| |
Collapse
|
48
|
Tamura T, Aoyama M, Ukai S, Kakita H, Sobue K, Asai K. Neuroprotective erythropoietin attenuates microglial activation, including morphological changes, phagocytosis, and cytokine production. Brain Res 2017; 1662:65-74. [PMID: 28257780 DOI: 10.1016/j.brainres.2017.02.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/27/2017] [Accepted: 02/21/2017] [Indexed: 12/12/2022]
Abstract
Erythropoietin (EPO), a hematopoietic hormonal cytokine induced in response to hypoxia, has neuroprotective effects. EPO receptor (EPOR) is expressed in microglia, resident immune cells in the brain. However, the effect of EPO on microglial activation is not clear. In the present study, we demonstrated that the EPOR is highly expressed in microglia, rather than in neurons or astrocytes, in in vitro experiments. Therefore, we investigated whether EPO could attenuate lipopolysaccharide (LPS)-mediated activation of microglia in vitro. The BV-2 microglial cell line was treated with LPS in the absence or presence of EPO. In the presence of EPO, microglial expression of LPS-induced inflammatory cytokine genes was significantly decreased. In addition, EPO suppressed the LPS-induced phagocytic activity of BV-2 cells towards fluorescent beads, as well as induction of inducible nitric oxide synthase. In in vivo experiments, EPO significantly decreased the LPS-induced expression of inflammatory cytokine genes in mouse brains. Furthermore, morphological analysis of cortical microglia in the brains of mice stimulated with LPS revealed that combined treatment with EPO alleviated LPS-induced morphological changes in the microglia. These data indicate that EPO attenuates microglial activation, including morphological changes in vivo, phagocytosis in vitro, and the production of inflammatory cytokines in vivo and in vitro. Further investigation of EPO modulation of LPS-induced microglial activation may contribute to the development of novel neuroprotective therapies.
Collapse
Affiliation(s)
- Tetsuya Tamura
- Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; Department of Anesthesiology and Intensive Care Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Mineyoshi Aoyama
- Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, Nagoya, Japan.
| | - Seiko Ukai
- Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Hiroki Kakita
- Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; Department of Perinatal and Neonatal Medicine, Aichi Medical University, Nagakute, Japan.
| | - Kazuya Sobue
- Department of Anesthesiology and Intensive Care Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Kiyofumi Asai
- Department of Molecular Neurobiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| |
Collapse
|
49
|
George J, Cunha RA, Mulle C, Amédée T. Microglia-derived purines modulate mossy fibre synaptic transmission and plasticity through P2X4 and A1 receptors. Eur J Neurosci 2016; 43:1366-78. [PMID: 27199162 PMCID: PMC5069607 DOI: 10.1111/ejn.13191] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/21/2016] [Indexed: 12/15/2022]
Abstract
Recent data have provided evidence that microglia, the brain‐resident macrophage‐like cells, modulate neuronal activity in both physiological and pathophysiological conditions, and microglia are therefore now recognized as synaptic partners. Among different neuromodulators, purines, which are produced and released by microglia, have emerged as promising candidates to mediate interactions between microglia and synapses. The cellular effects of purines are mediated through a large family of receptors for adenosine and for ATP (P2 receptors). These receptors are present at brain synapses, but it is unknown whether they can respond to microglia‐derived purines to modulate synaptic transmission and plasticity. Here, we used a simple model of adding immune‐challenged microglia to mouse hippocampal slices to investigate their impact on synaptic transmission and plasticity at hippocampal mossy fibre (MF) synapses onto CA3 pyramidal neurons. MF–CA3 synapses show prominent forms of presynaptic plasticity that are involved in the encoding and retrieval of memory. We demonstrate that microglia‐derived ATP differentially modulates synaptic transmission and short‐term plasticity at MF–CA3 synapses by acting, respectively, on presynaptic P2X4 receptors and on adenosine A1 receptors after conversion of extracellular ATP to adenosine. We also report that P2X4 receptors are densely located in the mossy fibre tract in the dentate gyrus–CA3 circuitry. In conclusion, this study reveals an interplay between microglia‐derived purines and MF–CA3 synapses, and highlights microglia as potent modulators of presynaptic plasticity.
Collapse
Affiliation(s)
- Jimmy George
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, University of Bordeaux, Bordeaux, France.,CNC Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, University of Bordeaux, Bordeaux, France
| | - Thierry Amédée
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, University of Bordeaux, Bordeaux, France.,IINS, UMR 5297 CNRS - Université de Bordeaux, Bordeaux Cedex, France
| |
Collapse
|
50
|
Ma Y, Wang J, Wang Y, Yang GY. The biphasic function of microglia in ischemic stroke. Prog Neurobiol 2016; 157:247-272. [PMID: 26851161 DOI: 10.1016/j.pneurobio.2016.01.005] [Citation(s) in RCA: 539] [Impact Index Per Article: 59.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/22/2015] [Accepted: 01/10/2016] [Indexed: 12/16/2022]
Abstract
Microglia are brain resident macrophages originated from primitive progenitor cells in the yolk sac. Microglia can be activated within hours and recruited to the lesion site. Traditionally, microglia activation is considered to play a deleterious role in ischemic stroke, as inhibition of microglia activation attenuates ischemia induced brain injury. However, increasing evidence show that microglia activation is critical for attenuating neuronal apoptosis, enhancing neurogenesis, and promoting functional recovery after cerebral ischemia. Differential polarization of microglia could likely explain the biphasic role of microglia in ischemia. We comprehensively reviewed the mechanisms involved in regulating microglia activation and polarization. The latest discoveries of microRNAs in modulating microglia function are discussed. In addition, the interaction between microglia and other cells including neurons, astrocytes, oligodendrocytes, and stem cells were also reviewed. Future therapies targeting microglia may not exclusively aim at suppressing microglia activation, but also at modulating microglia polarization at different stages of ischemic stroke. More work is needed to elucidate the cellular and molecular mechanisms of microglia polarization under ischemic environment. The roles of microRNAs and transplanted stem cells in mediating microglia activation and polarization during brain ischemia also need to be further studied.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jixian Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Rehabilitation, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|